1
|
Treves N, Yakirevich-Amir N, Abu Ahmad W, Bonne O, Davidson E, Keeling K, Hall B, Dautrich T, Matok I. Characterization of Cannabis users and products and the experience of negative mental emotions following Cannabis use. Eur Arch Psychiatry Clin Neurosci 2025; 275:407-420. [PMID: 38861239 PMCID: PMC11910409 DOI: 10.1007/s00406-024-01812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/19/2024] [Indexed: 06/12/2024]
Abstract
There is a potential link between cannabis and mental disorders. Cannabis exposure involves in many cases negative mental emotions, which are unpleasant sensations or thoughts. Whereas mild cases of negative mental emotions inflict patient's quality of life, more severe cases lead to therapy discontinuations, or even hospitalizations and death. This study characterizes cannabis users who experienced negative mental emotions after cannabis exposure. The Releaf App database was utilized to evaluate the association between personal and cannabis use characteristics on reporting a negative mental emotion during cannabis exposure. This global mobile lets individuals track real-time cannabis experience use with cannabinoid-based products, containing data points such as gender, age, reasons for use, product type, cannabis composition, and feelings and emotions experienced after cannabis use. Multivariable logistic regression models were constructed, adjusting for potential confounders such as gender and previous experience with cannabis use. The study population comprised 4,435 users, and 34,279 sessions were collected from various countries, mainly from North America, and included in the primary analysis. Reporting on negative mental emotions was associated with users in the age group of 18-30 years. Using cannabis for a mental purpose was associated with a small increase in reporting on negative mental emotions (OR = 1.10, 95%CI [1.03-1.19]). Oral products were associated with reporting on negative mental emotions. THC-dominant products were associated with reporting negative mental emotions compared to balanced products (OR = 1.21, 95%CI [1.06-1.38]). This study suggests that some characteristics of cannabis use, such as young age and oral consumption are associated with negative mental emotions. Further studies should examine the interface between cannabis consumption, characteristics of consumers, and negative emotional experience or even long-term mental disorders.
Collapse
Affiliation(s)
- Nir Treves
- Division of Clinical Pharmacy, School of Pharmacy, The Hebrew University, Jerusalem, Israel.
| | - Noa Yakirevich-Amir
- Department of Psychiatry, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Wiessam Abu Ahmad
- Hebrew University, Hadassah Braun School of Public Health and Community Medicine, Jerusalem, Israel
| | - Omer Bonne
- Department of Psychiatry, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Elyad Davidson
- Department of Anesthesia CCM and Pain Management, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | - Branden Hall
- MoreBetter, Ltd. (dba Releaf App), Hyattsville, MD, USA
| | | | - Ilan Matok
- Division of Clinical Pharmacy, School of Pharmacy, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
2
|
Kumar U. Cannabinoids: Role in Neurological Diseases and Psychiatric Disorders. Int J Mol Sci 2024; 26:152. [PMID: 39796008 PMCID: PMC11720483 DOI: 10.3390/ijms26010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
An impact of legalization and decriminalization of marijuana is the gradual increase in the use of cannabis for recreational purposes, which poses a potential threat to society and healthcare systems worldwide. However, the discovery of receptor subtypes, endogenous endocannabinoids, and enzymes involved in synthesis and degradation, as well as pharmacological characterization of receptors, has led to exploration of the use of cannabis in multiple peripheral and central pathological conditions. The role of cannabis in the modulation of crucial events involving perturbed physiological functions and disease progression, including apoptosis, inflammation, oxidative stress, perturbed mitochondrial function, and the impaired immune system, indicates medicinal values. These events are involved in most neurological diseases and prompt the gradual progression of the disease. At present, several synthetic agonists and antagonists, in addition to more than 70 phytocannabinoids, are available with distinct efficacy as a therapeutic alternative in different pathological conditions. The present review aims to describe the use of cannabis in neurological diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
3
|
Wang Z, Arnold JC. Cannabinoids and healthy ageing: the potential for extending healthspan and lifespan in preclinical models with an emphasis on Caenorhabditis elegans. GeroScience 2024; 46:5643-5661. [PMID: 38696056 PMCID: PMC11493940 DOI: 10.1007/s11357-024-01162-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/11/2024] [Indexed: 10/23/2024] Open
Abstract
There is a significant global upsurge in the number and proportion of older persons in the population. With this comes an increasing prevalence of age-related conditions which pose a major challenge to healthcare systems. The development of anti-ageing treatments may help meet this challenge by targeting the ageing process which is a common denominator to many health problems. Cannabis-like compounds (cannabinoids) are reported to improve quality of life and general well-being in human trials, and there is increasing preclinical research highlighting that they have anti-ageing activity. Moreover, preclinical evidence suggests that endogenous cannabinoids regulate ageing processes. Here, we review the anti-ageing effects of the cannabinoids in various model systems, including the most extensively studied nematode model, Caenorhabditis elegans. These studies highlight that the cannabinoids lengthen healthspan and lifespan, with emerging evidence that they may also hinder the development of cellular senescence. The non-psychoactive cannabinoid cannabidiol (CBD) shows particular promise, with mechanistic studies demonstrating it may work through autophagy induction and activation of antioxidative systems. Furthermore, CBD improves healthspan parameters such as diminishing age-related behavioural dysfunction in models of both healthy and accelerated ageing. Translation into mammalian systems provides an important next step. Moreover, looking beyond CBD, future studies could probe the multitude of other cannabis constituents for their anti-ageing activity.
Collapse
Affiliation(s)
- Zhizhen Wang
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Jonathon C Arnold
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
- Discipline of Pharmacology, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Bilkei-Gorzo A, Schurmann B, Schneider M, Kraemer M, Nidadavolu P, Beins EC, Müller CE, Dvir-Ginzberg M, Zimmer A. Bidirectional Effect of Long-Term Δ 9-Tetrahydrocannabinol Treatment on mTOR Activity and Metabolome. ACS Pharmacol Transl Sci 2024; 7:2637-2649. [PMID: 39296258 PMCID: PMC11406684 DOI: 10.1021/acsptsci.4c00002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/15/2024] [Accepted: 06/18/2024] [Indexed: 09/21/2024]
Abstract
Brain aging is associated with cognitive decline, reduced synaptic plasticity, and altered metabolism. The activity of mechanistic target of rapamycin (mTOR) has a major impact on aging by regulating cellular metabolism. Although reduced mTOR signaling has a general antiaging effect, it can negatively affect the aging brain by reducing synaptogenesis and thus cognitive functions. Increased mTOR activity facilitates aging and is responsible for the amnestic effect of the cannabinoid receptor 1 agonist Δ9-tetrahydrocannabinol (THC) in higher doses. Long-term low-dose Δ9-THC had an antiaging effect on the brain by restoring cognitive abilities and synapse densities in old mice. Whether changes in mTOR signaling and metabolome are associated with its positive effects on the aging brain is an open question. Here, we show that Δ9-THC treatment has a tissue-dependent and dual effect on mTOR signaling and the metabolome. In the brain, Δ9-THC treatment induced a transient increase in mTOR activity and in the levels of amino acids and metabolites involved in energy production, followed by an increased synthesis of synaptic proteins. Unexpectedly, we found a similar reduction in the mTOR activity in adipose tissue and in the level of amino acids and carbohydrate metabolites in blood plasma as in animals on a low-calorie diet. Thus, long-term Δ9-THC treatment first increases the level of energy and synaptic protein production in the brain, followed by a reduction in mTOR activity and metabolic processes in the periphery. Our study suggests that a dual effect on mTOR activity and the metabolome could be the basis for an effective antiaging and pro-cognitive medication.
Collapse
Affiliation(s)
- Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Britta Schurmann
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Marion Schneider
- Pharmaceutical Institute, University of Bonn, Bonn 53121, Germany
| | - Michael Kraemer
- Institute of Forensic Medicine, Medical Faculty, University of Bonn, Bonn 53111, Germany
| | - Prakash Nidadavolu
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Eva C Beins
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Christa E Müller
- Pharmaceutical Institute, University of Bonn, Bonn 53121, Germany
| | - Mona Dvir-Ginzberg
- Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| |
Collapse
|
5
|
Meng H, Sengupta A, Ricciotti E, Mrčela A, Mathew D, Mazaleuskaya LL, Ghosh S, Brooks TG, Turner AP, Schanoski AS, Lahens NF, Tan AW, Woolfork A, Grant G, Susztak K, Letizia AG, Sealfon SC, Wherry EJ, Laudanski K, Weljie AM, Meyer NJ, FitzGerald GA. Deep phenotyping of the lipidomic response in COVID-19 and non-COVID-19 sepsis. Clin Transl Med 2023; 13:e1440. [PMID: 37948331 PMCID: PMC10637636 DOI: 10.1002/ctm2.1440] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/15/2023] [Accepted: 10/01/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Lipids may influence cellular penetrance by viral pathogens and the immune response that they evoke. We deeply phenotyped the lipidomic response to SARs-CoV-2 and compared that with infection with other pathogens in patients admitted with acute respiratory distress syndrome to an intensive care unit (ICU). METHODS Mass spectrometry was used to characterise lipids and relate them to proteins, peripheral cell immunotypes and disease severity. RESULTS Circulating phospholipases (sPLA2, cPLA2 (PLA2G4A) and PLA2G2D) were elevated on admission in all ICU groups. Cyclooxygenase, lipoxygenase and epoxygenase products of arachidonic acid (AA) were elevated in all ICU groups compared with controls. sPLA2 predicted severity in COVID-19 and correlated with TxA2, LTE4 and the isoprostane, iPF2α-III, while PLA2G2D correlated with LTE4. The elevation in PGD2, like PGI2 and 12-HETE, exhibited relative specificity for COVID-19 and correlated with sPLA2 and the interleukin-13 receptor to drive lymphopenia, a marker of disease severity. Pro-inflammatory eicosanoids remained correlated with severity in COVID-19 28 days after admission. Amongst non-COVID ICU patients, elevations in 5- and 15-HETE and 9- and 13-HODE reflected viral rather than bacterial disease. Linoleic acid (LA) binds directly to SARS-CoV-2 and both LA and its di-HOME products reflected disease severity in COVID-19. In healthy marines, these lipids rose with seroconversion. Eicosanoids linked variably to the peripheral cellular immune response. PGE2, TxA2 and LTE4 correlated with T cell activation, as did PGD2 with non-B non-T cell activation. In COVID-19, LPS stimulated peripheral blood mononuclear cell PGF2α correlated with memory T cells, dendritic and NK cells while LA and DiHOMEs correlated with exhausted T cells. Three high abundance lipids - ChoE 18:3, LPC-O-16:0 and PC-O-30:0 - were altered specifically in COVID. LPC-O-16:0 was strongly correlated with T helper follicular cell activation and all three negatively correlated with multi-omic inflammatory pathways and disease severity. CONCLUSIONS A broad based lipidomic storm is a predictor of poor prognosis in ARDS. Alterations in sPLA2, PGD2 and 12-HETE and the high abundance lipids, ChoE 18:3, LPC-O-16:0 and PC-O-30:0 exhibit relative specificity for COVID-19 amongst such patients and correlate with the inflammatory response to link to disease severity.
Collapse
Affiliation(s)
- Hu Meng
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Emanuela Ricciotti
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Antonijo Mrčela
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Divij Mathew
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Immunology and Immune HealthPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Liudmila L. Mazaleuskaya
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Soumita Ghosh
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Thomas G. Brooks
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Alexandra P. Turner
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Nicholas F. Lahens
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ai Wen Tan
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ashley Woolfork
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Greg Grant
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of GeneticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Katalin Susztak
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Andrew G. Letizia
- Naval Medical Research CenterSilver SpringMarylandUSA
- Naval Medical Research Unit TWOSingaporeSingapore
| | - Stuart C. Sealfon
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - E. John Wherry
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Immunology and Immune HealthPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Critical CarePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Aalim M. Weljie
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Nuala J. Meyer
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Garret A. FitzGerald
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
6
|
Maccarrone M, Di Marzo V, Gertsch J, Grether U, Howlett AC, Hua T, Makriyannis A, Piomelli D, Ueda N, van der Stelt M. Goods and Bads of the Endocannabinoid System as a Therapeutic Target: Lessons Learned after 30 Years. Pharmacol Rev 2023; 75:885-958. [PMID: 37164640 PMCID: PMC10441647 DOI: 10.1124/pharmrev.122.000600] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023] Open
Abstract
The cannabis derivative marijuana is the most widely used recreational drug in the Western world and is consumed by an estimated 83 million individuals (∼3% of the world population). In recent years, there has been a marked transformation in society regarding the risk perception of cannabis, driven by its legalization and medical use in many states in the United States and worldwide. Compelling research evidence and the Food and Drug Administration cannabis-derived cannabidiol approval for severe childhood epilepsy have confirmed the large therapeutic potential of cannabidiol itself, Δ9-tetrahydrocannabinol and other plant-derived cannabinoids (phytocannabinoids). Of note, our body has a complex endocannabinoid system (ECS)-made of receptors, metabolic enzymes, and transporters-that is also regulated by phytocannabinoids. The first endocannabinoid to be discovered 30 years ago was anandamide (N-arachidonoyl-ethanolamine); since then, distinct elements of the ECS have been the target of drug design programs aimed at curing (or at least slowing down) a number of human diseases, both in the central nervous system and at the periphery. Here a critical review of our knowledge of the goods and bads of the ECS as a therapeutic target is presented to define the benefits of ECS-active phytocannabinoids and ECS-oriented synthetic drugs for human health. SIGNIFICANCE STATEMENT: The endocannabinoid system plays important roles virtually everywhere in our body and is either involved in mediating key processes of central and peripheral diseases or represents a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of the components of this complex system, and in particular of key receptors (like cannabinoid receptors 1 and 2) and metabolic enzymes (like fatty acid amide hydrolase and monoacylglycerol lipase), will advance our understanding of endocannabinoid signaling and activity at molecular, cellular, and system levels, providing new opportunities to treat patients.
Collapse
Affiliation(s)
- Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Vincenzo Di Marzo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Jürg Gertsch
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Uwe Grether
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Allyn C Howlett
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Tian Hua
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Alexandros Makriyannis
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Daniele Piomelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Natsuo Ueda
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Mario van der Stelt
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| |
Collapse
|
7
|
Lee Y, Kim Y, Park S, Heo G, Chung HY, Im E. Cannabinoid receptor type 1 in the aging gut regulates the mucosal permeability via miR-191-5p. Front Endocrinol (Lausanne) 2023; 14:1241097. [PMID: 37693348 PMCID: PMC10485608 DOI: 10.3389/fendo.2023.1241097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Background Aging is associated with a broad loss of function throughout the body, and gastrointestinal (GI) dysfunction can occur with aging. The endocannabinoid (eCB) system plays a pivotal role in various GI diseases, and alterations in the eCB system have been observed during brain and skin aging. Therefore, we investigated the putative role of the eCB system in aging-related changes in the intestine. Methods The expression of cannabinoid receptor type 1 (CB1) was investigated in rat intestinal tissues using quantitative real-time PCR. Cellular senescence was induced by hydrogen peroxide (H2O2) and hydroxyurea (HU) in rat and human intestinal epithelial cells. Cellular permeability was evaluated by transepithelial electrical resistance (TEER) measurement. Results and Discussion The expression of CB1 was decreased in the small intestine of aged rats compared to that of young rats. Senescent cells showed reduced TEER values and decreased expression of ZO-1, indicating increased intestinal permeability, which is tightly regulated by the CB1 signaling. In silico miRNA analysis suggested that ZO-1 was a direct target gene of miR-191-5p. Increased expression of miR-191-5p by HU was restored by CB1 agonist ACEA co-treatment. Moreover, NF-κB p65 activation was associated with CB1-related miR-191-5p signaling. In conclusion, aging-induced CB1 reduction leads to increased intestinal permeability and decreased ZO-1 expression via upregulation of miR-191-5p and NF-κB p65 activation. Taken together, these results suggest that CB1 signaling may be a useful strategy to reduce intestinal permeability in aging-related and other inflammatory conditions in the gut.
Collapse
Affiliation(s)
- Yunna Lee
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Yuju Kim
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Soyeong Park
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Gwangbeom Heo
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Hae Young Chung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
- Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
8
|
Meng H, Sengupta A, Ricciotti E, Mrčela A, Mathew D, Mazaleuskaya LL, Ghosh S, Brooks TG, Turner AP, Schanoski AS, Lahens NF, Tan AW, Woolfork A, Grant G, Susztak K, Letizia AG, Sealfon SC, Wherry EJ, Laudanski K, Weljie AM, Meyer NB, FitzGerald GA. Deep Phenotyping of the Lipidomic Response in COVID and non-COVID Sepsis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543298. [PMID: 37398323 PMCID: PMC10312560 DOI: 10.1101/2023.06.02.543298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lipids may influence cellular penetrance by pathogens and the immune response that they evoke. Here we find a broad based lipidomic storm driven predominantly by secretory (s) phospholipase A 2 (sPLA 2 ) dependent eicosanoid production occurs in patients with sepsis of viral and bacterial origin and relates to disease severity in COVID-19. Elevations in the cyclooxygenase (COX) products of arachidonic acid (AA), PGD 2 and PGI 2 , and the AA lipoxygenase (LOX) product, 12-HETE, and a reduction in the high abundance lipids, ChoE 18:3, LPC-O-16:0 and PC-O-30:0 exhibit relative specificity for COVID-19 amongst such patients, correlate with the inflammatory response and link to disease severity. Linoleic acid (LA) binds directly to SARS-CoV-2 and both LA and its di-HOME products reflect disease severity in COVID-19. AA and LA metabolites and LPC-O-16:0 linked variably to the immune response. These studies yield prognostic biomarkers and therapeutic targets for patients with sepsis, including COVID-19. An interactive purpose built interactive network analysis tool was developed, allowing the community to interrogate connections across these multiomic data and generate novel hypotheses.
Collapse
|
9
|
Palmisano M, Gargano A, Olabiyi BF, Lutz B, Bilkei-Gorzo A. Hippocampal Deletion of CB1 Receptor Impairs Social Memory and Leads to Age-Related Changes in the Hippocampus of Adult Mice. Int J Mol Sci 2022; 24:ijms24010026. [PMID: 36613469 PMCID: PMC9819823 DOI: 10.3390/ijms24010026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Endocannabinoid system activity declines with age in the hippocampus, along with the density of the cannabinoid receptor type-1 (CB1). This process might contribute to brain ageing, as previous studies showed that the constitutive deletion of the CB1 receptor in mice leads to early onset of memory deficits and histological signs of ageing in the hippocampus including enhanced pro-inflammatory glial activity and reduced neurogenesis. Here we asked whether the CB1 receptor exerts its activity locally, directly influencing hippocampal ageing or indirectly, accelerating systemic ageing. Thus, we deleted the CB1 receptor site-specifically in the hippocampus of 2-month-old CB1flox/flox mice using stereotaxic injections of rAAV-Cre-Venus viruses and assessed their social recognition memory four months later. Mice with hippocampus-specific deletion of the CB1 receptor displayed a memory impairment, similarly as observed in constitutive knockouts at the same age. We next analysed neuroinflammatory changes in the hippocampus, neuronal density and cell proliferation. Site-specific mutant mice had enhanced glial cell activity, up-regulated levels of TNFα in the hippocampus and decreased cell proliferation, specifically in the subgranular zone of the dentate gyrus. Our data indicate that a local activity of the CB1 receptor in the hippocampus is required to maintain neurogenesis and to prevent neuroinflammation and cognitive decline.
Collapse
Affiliation(s)
- Michela Palmisano
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53125 Bonn, Germany
| | - Alessandra Gargano
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53125 Bonn, Germany
| | | | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53125 Bonn, Germany
- Correspondence: ; Tel.: +49-0228-6885-317
| |
Collapse
|
10
|
Dohnalová L, Lundgren P, Carty JRE, Goldstein N, Wenski SL, Nanudorn P, Thiengmag S, Huang KP, Litichevskiy L, Descamps HC, Chellappa K, Glassman A, Kessler S, Kim J, Cox TO, Dmitrieva-Posocco O, Wong AC, Allman EL, Ghosh S, Sharma N, Sengupta K, Cornes B, Dean N, Churchill GA, Khurana TS, Sellmyer MA, FitzGerald GA, Patterson AD, Baur JA, Alhadeff AL, Helfrich EJN, Levy M, Betley JN, Thaiss CA. A microbiome-dependent gut-brain pathway regulates motivation for exercise. Nature 2022; 612:739-747. [PMID: 36517598 PMCID: PMC11162758 DOI: 10.1038/s41586-022-05525-z] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/04/2022] [Indexed: 12/16/2022]
Abstract
Exercise exerts a wide range of beneficial effects for healthy physiology1. However, the mechanisms regulating an individual's motivation to engage in physical activity remain incompletely understood. An important factor stimulating the engagement in both competitive and recreational exercise is the motivating pleasure derived from prolonged physical activity, which is triggered by exercise-induced neurochemical changes in the brain. Here, we report on the discovery of a gut-brain connection in mice that enhances exercise performance by augmenting dopamine signalling during physical activity. We find that microbiome-dependent production of endocannabinoid metabolites in the gut stimulates the activity of TRPV1-expressing sensory neurons and thereby elevates dopamine levels in the ventral striatum during exercise. Stimulation of this pathway improves running performance, whereas microbiome depletion, peripheral endocannabinoid receptor inhibition, ablation of spinal afferent neurons or dopamine blockade abrogate exercise capacity. These findings indicate that the rewarding properties of exercise are influenced by gut-derived interoceptive circuits and provide a microbiome-dependent explanation for interindividual variability in exercise performance. Our study also suggests that interoceptomimetic molecules that stimulate the transmission of gut-derived signals to the brain may enhance the motivation for exercise.
Collapse
Affiliation(s)
- Lenka Dohnalová
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Molecular Bio Science, Goethe University Frankfurt, and LOEWE Center for Translational Biodiversity Genomics, Frankfurt, Germany
| | - Patrick Lundgren
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jamie R E Carty
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nitsan Goldstein
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sebastian L Wenski
- Institute for Molecular Bio Science, Goethe University Frankfurt, and LOEWE Center for Translational Biodiversity Genomics, Frankfurt, Germany
| | - Pakjira Nanudorn
- Institute for Molecular Bio Science, Goethe University Frankfurt, and LOEWE Center for Translational Biodiversity Genomics, Frankfurt, Germany
| | - Sirinthra Thiengmag
- Institute for Molecular Bio Science, Goethe University Frankfurt, and LOEWE Center for Translational Biodiversity Genomics, Frankfurt, Germany
| | | | - Lev Litichevskiy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hélène C Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karthikeyani Chellappa
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana Glassman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susanne Kessler
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jihee Kim
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy O Cox
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Oxana Dmitrieva-Posocco
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea C Wong
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erik L Allman
- Department of Biochemistry and Molecular Biology and Department of Veterinary and Biomedical Sciences, the Pennsylvania State University, University Park, PA, USA
| | - Soumita Ghosh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nitika Sharma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kasturi Sengupta
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Tejvir S Khurana
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D Patterson
- Department of Biochemistry and Molecular Biology and Department of Veterinary and Biomedical Sciences, the Pennsylvania State University, University Park, PA, USA
| | - Joseph A Baur
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amber L Alhadeff
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric J N Helfrich
- Institute for Molecular Bio Science, Goethe University Frankfurt, and LOEWE Center for Translational Biodiversity Genomics, Frankfurt, Germany
| | - Maayan Levy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J Nicholas Betley
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Radhakrishnan R, Worhunsky PD, Zheng MQ, Najafzadeh S, Gallezot JD, Planeta B, Henry S, Nabulsi N, Ranganathan M, Skosnik PD, Pittman B, Cyril D'Souza D, Carson RE, Huang Y, Potenza MN, Matuskey D. Age, gender and body-mass-index relationships with in vivo CB 1 receptor availability in healthy humans measured with [ 11C]OMAR PET. Neuroimage 2022; 264:119674. [PMID: 36243269 DOI: 10.1016/j.neuroimage.2022.119674] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 09/07/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
Brain cannabinoid 1 receptors (CB1Rs) contribute importantly to the regulation of autonomic tone, appetite, mood and cognition. Inconsistent results have been reported from positron emission tomography (PET) studies using different radioligands to examine relationships between age, gender and body mass index (BMI) and CB1R availability in healthy individuals. In this study, we examined these variables in 58 healthy individuals (age range: 18-55 years; 44 male; BMI=27.01±5.56), the largest cohort of subjects studied to date using the CB1R PET ligand [11C]OMAR. There was a significant decline in CB1R availability (VT) with age in the pallidum, cerebellum and posterior cingulate. Adjusting for BMI, age-related decline in VT remained significant in the posterior cingulate among males, and in the cerebellum among women. CB1R availability was higher in women compared to men in the thalamus, pallidum and posterior cingulate. Adjusting for age, CB1R availability negatively correlated with BMI in women but not men. These findings differ from those reported using [11C]OMAR and other radioligands such as [18F]FMPEP-d2 and [18F]MK-9470. Although reasons for these seemingly divergent findings are unclear, the choice of PET radioligand and range of BMI in the current dataset may contribute to the observed differences. This study highlights the need for cross-validation studies using both [11C]OMAR and [18F]FMPEP-d2 within the same cohort of subjects.
Collapse
Affiliation(s)
- Rajiv Radhakrishnan
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States.
| | - Patrick D Worhunsky
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States
| | - Ming-Qiang Zheng
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Soheila Najafzadeh
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Beata Planeta
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Shannan Henry
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Mohini Ranganathan
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States
| | - Patrick D Skosnik
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States
| | - Brian Pittman
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States
| | - Deepak Cyril D'Souza
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States; Child Study Center, Yale University School of Medicine, United States; Connecticut Mental Health Center, United States; Department of Neuroscience, Yale University, United States
| | - David Matuskey
- Department of Psychiatry, Yale University School of Medicinev, New Haven, CT 06511, United States; Department of Radiology and Biomedical Imaging, Yale University School of Medicine, United States; Department of Neurology, Yale University School of Medicine, United States
| |
Collapse
|
12
|
Cherkasova V, Wang B, Gerasymchuk M, Fiselier A, Kovalchuk O, Kovalchuk I. Use of Cannabis and Cannabinoids for Treatment of Cancer. Cancers (Basel) 2022; 14:5142. [PMID: 36291926 PMCID: PMC9600568 DOI: 10.3390/cancers14205142] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 07/26/2023] Open
Abstract
The endocannabinoid system (ECS) is an ancient homeostasis mechanism operating from embryonic stages to adulthood. It controls the growth and development of many cells and cell lineages. Dysregulation of the components of the ECS may result in uncontrolled proliferation, adhesion, invasion, inhibition of apoptosis and increased vascularization, leading to the development of various malignancies. Cancer is the disease of uncontrolled cell division. In this review, we will discuss whether the changes to the ECS are a cause or a consequence of malignization and whether different tissues react differently to changes in the ECS. We will discuss the potential use of cannabinoids for treatment of cancer, focusing on primary outcome/care-tumor shrinkage and eradication, as well as secondary outcome/palliative care-improvement of life quality, including pain, appetite, sleep, and many more factors. Finally, we will complete this review with the chapter on sex- and gender-specific differences in ECS and response to cannabinoids, and equality of the access to treatments with cannabinoids.
Collapse
Affiliation(s)
- Viktoriia Cherkasova
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Marta Gerasymchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Anna Fiselier
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
13
|
Dynamic Changes in the Endocannabinoid System during the Aging Process: Focus on the Middle-Age Crisis. Int J Mol Sci 2022; 23:ijms231810254. [PMID: 36142165 PMCID: PMC9499672 DOI: 10.3390/ijms231810254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Endocannabinoid (eCB) signaling is markedly decreased in the hippocampus (Hip) of aged mice, and the genetic deletion of the cannabinoid receptor type 1 (CB1) leads to an early onset of cognitive decline and age-related histological changes in the brain. Thus, it is hypothesized that cognitive aging is modulated by eCB signaling through CB1. In the present study, we detailed the changes in the eCB system during the aging process using different complementary techniques in mouse brains of five different age groups, ranging from adolescence to old age. Our findings indicate that the eCB system is most strongly affected in middle-aged mice (between 9 and 12 months of age) in a brain region-specific manner. We show that 2-arachidonoylglycerol (2-AG) was prominently decreased in the Hip and moderately in caudate putamen (CPu), whereas anandamide (AEA) was decreased in both CPu and medial prefrontal cortex along with cingulate cortex (mPFC+Cg), starting from 6 months until 12 months. Consistent with the changes in 2-AG, the 2-AG synthesizing enzyme diacylglycerol lipase α (DAGLα) was also prominently decreased across the sub-regions of the Hip. Interestingly, we found a transient increase in CB1 immunoreactivity across the sub-regions of the Hip at 9 months, a plausible compensation for reduced 2-AG, which ultimately decreased strongly at 12 months. Furthermore, quantitative autoradiography of CB1 revealed that [3H]CP55940 binding markedly increased in the Hip at 9 months. However, unlike the protein levels, CB1 binding density did not drop strongly at 12 months and at old age. Furthermore, [3H]CP55940 binding was significantly increased in the lateral entorhinal cortex (LEnt), starting from the middle age until the old age. Altogether, our findings clearly indicate a middle-age crisis in the eCB system, which could be a potential time window for therapeutic interventions to abrogate the course of cognitive aging.
Collapse
|
14
|
Sloan ME, Grant CW, Stangl BL, Klepp TD, Brewton HW, Cinar R, Kunos G, Ramchandani VA. The effects of acute alcohol administration on circulating endocannabinoid levels in humans. Addict Biol 2022; 27:e13197. [PMID: 36001429 PMCID: PMC9413364 DOI: 10.1111/adb.13197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 04/26/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022]
Abstract
Several lines of evidence suggest that endocannabinoid signalling may influence alcohol consumption. Preclinical studies have found that pharmacological blockade of cannabinoid receptor 1 leads to reductions in alcohol intake. Furthermore, variations in endocannabinoid metabolism between individuals may be associated with the presence and severity of alcohol use disorder. However, little is known about the acute effects of alcohol on the endocannabinoid system in humans. In this study, we evaluated the effect of acute alcohol administration on circulating endocannabinoid levels by analysing data from two highly-controlled alcohol administration experiments. In the first within-subjects experiment, 47 healthy participants were randomized to receive alcohol and placebo in a counterbalanced order. Alcohol was administered using an intravenous clamping procedure such that each participant attained a nearly identical breath alcohol concentration of 0.05%, maintained over 3 h. In the second experiment, 23 healthy participants self-administered alcohol intravenously; participants had control over their exposure throughout the paradigm. In both experiments, circulating concentrations of two endocannabinoids, N-arachidonoylethanolamine (AEA) and 2-arachidonoylglycerol (2-AG), were measured at baseline and following alcohol exposure. During the intravenous clamping procedure, acute alcohol administration reduced circulating AEA but not 2-AG levels when compared to placebo. This finding was confirmed in the self-administration paradigm, where alcohol reduced AEA levels in an exposure-dependent manner. Future studies should seek to determine whether alcohol administration has similar effects on brain endocannabinoid signalling. An improved understanding of the bidirectional relationship between endocannabinoid signalling and alcohol intake may deepen our understanding of the aetiology and repercussions of alcohol use disorder.
Collapse
Affiliation(s)
- Matthew E Sloan
- Addictions Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Division of Neurosciences and Clinical Translation, Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychological Clinical Science, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Caroline W Grant
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Bethany L Stangl
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Timothy D Klepp
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Honoree W Brewton
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Resat Cinar
- Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Vijay A Ramchandani
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
15
|
Sánchez-Marín L, Javier Pavón-Morón F, Rodríguez de Fonseca F, Serrano A. Attenuation of Oleoylethanolamide-Induced Reduction of Alcohol Consumption in Adult Rats Exposed Intermittently to Alcohol During Adolescence. Neurosci Lett 2022; 781:136670. [DOI: 10.1016/j.neulet.2022.136670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/15/2022]
|
16
|
Acute stress and alcohol exposure during adolescence result in an anxious phenotype in adulthood: Role of altered glutamate/endocannabinoid transmission mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 2022; 113:110460. [PMID: 34695542 DOI: 10.1016/j.pnpbp.2021.110460] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Stressful episodes and high alcohol consumption during adolescence are considered major risk factors for the development of psychiatric disorders in adulthood. Identification of mechanisms underlying these early events, which enhanced vulnerability to mental illness, is essential for both their prevention and treatment. METHODS Male Wistar rats were used to investigate the long-term effects of early restraint stress and intermittent alcohol exposure (intragastric administration of 3 g/kg ethanol; 4 days/week for 4 weeks during adolescence) on anxiety-like behavior and the expression of signaling systems associated with emotional behaviors [e.g., corticosterone, fatty acid-derived molecules and endocannabinoid enzymes, glutamate receptor subunits, corticotropin releasing hormone receptors (CRHR1 and CRHR2) and neuropeptide Y receptors (NPY1R and NPYR2)] in the blood and amygdala. RESULTS Overall, both stress and alcohol exposure during adolescence induced anxiogenic-like behaviors, increased plasma levels of corticosterone and increases in the amygdalar expression of the cannabinoid CB2 receptor and certain subunits of glutamate receptors (i.e., mGluR1, mGluR5 and NMDAR1) in young adult rats. In addition, there were specific main effects of alcohol exposure on the expression of the cannabinoid CB1 receptor, monoacylglycerol lipase (MAGL) and NPY2R in the amygdala, and significant increases were observed in rats exposed to alcohol. Interestingly, there were significant interaction effects between restraint stress and alcohol exposure on the expression of plasma 2-arachidonoyl glycerol (2-AG), and both CRHR1,2 and NPY1R in the amygdala. Thus, the restraint stress was associated with increased 2-AG levels, which was not observed in rats exposed to alcohol. The alcohol exposure was associated with an increased expression of CRHR1,2 but the restraint stress prevented these increases (stress alcohol rats). In contrast, NPY1R was only increased in rats exposed to stress and alcohol. Finally, we did not observe any potentiation of the behavioral and molecular effects by the combination of stress and alcohol, which is concordant with an overall ceiling effect on some of the variables. CONCLUSION Separate and combined early stress and alcohol induced a common anxious phenotype with increased corticosterone in adulthood. However, there were differences in the amygdalar expression of signaling systems involved in maladaptive changes in emotional behavior. Therefore, our results suggest the existence of partially different mechanisms for stress and alcohol exposures.
Collapse
|
17
|
de Biedma-Elduayen LG, Giménez-Gómez P, Morales-Puerto N, Vidal R, de la Calle CN, Gutiérrez-López MD, O'Shea E, Colado MI. Influx of kynurenine into the brain is involved in the reduction of ethanol consumption induced by Ro 61-8048 after chronic intermittent ethanol in mice. Br J Pharmacol 2022; 179:3711-3726. [PMID: 35189673 PMCID: PMC9314579 DOI: 10.1111/bph.15825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/07/2022] [Accepted: 02/10/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The kynurenine pathway has been proposed as a new target for modulating drug abuse. We previously demonstrated that inhibition of kynurenine 3-monooxygenase (KMO) using Ro 61-8048 reduces ethanol consumption in a binge drinking model. Here we investigate the effect of the kynurenine pathway modulation in ethanol -dependent mice. EXPERIMENTAL APPROACH Adult male and female mice were subjected to the Chronic Intermittent Ethanol (CIE) paradigm. On the last day of CIE, mice were treated with Ro 61-8048, Ro 61-8048 + PNU-120596, a positive allosteric modulator of α7nAChR, and Ro 61-8048 + L-leucine or probenecid, which block the influx or efflux of kynurenine from the brain, respectively. Ethanol, water consumption and preference were measured and kynurenine levels in plasma and limbic forebrain were determined. KEY RESULTS Ro 61-8048 decreases consumption and preference for ethanol in both sexes exposed to the CIE model, an effect that is prevented by PNU-120596. The Ro 61-8048-induced decrease in ethanol consumption depends on the influx of kynurenine into the brain. CONCLUSION AND IMPLICATIONS Inhibition of KMO reduces ethanol consumption and preference in both male and female mice subjected to CIE model by a mechanism involving α7nAChR. Moreover, the effect which is mediated centrally depends on the influx of peripheral kynurenine to the brain and can be prolonged by blocking the efflux of kynurenine from the brain. Here, for the first time we demonstrate that the modulation of the kynurenine pathway is a valid strategy for the treatment of ethanol dependence in both sexes.
Collapse
Affiliation(s)
- Leticia Gil de Biedma-Elduayen
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Current address: University of Massachusetts Chan Medical School, The Brudnick Neuropsychiatric Research Institute, Worcester, MA
| | - Nuria Morales-Puerto
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Rebeca Vidal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Carlos Núñez de la Calle
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María Dolores Gutiérrez-López
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
18
|
Ginsburg BC, Hensler JG. Age-related changes in CB1 receptor expression and function and the behavioral effects of cannabinoid receptor ligands. Pharmacol Biochem Behav 2022; 213:173339. [PMID: 35077729 PMCID: PMC8973309 DOI: 10.1016/j.pbb.2022.173339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/03/2023]
Abstract
Cannabinoid use has increased among aging individuals. However, little information on age-related differences in the behavioral effects of these agents is available. To explore potential differences in the behavioral effects of cannabinoids, we determined effects of Δ9-tetrahydrocannabinol (THC, 1-10 mg/kg) or rimonabant (0.3-3.2 mg/kg) on operant fixed-ratio responding (FR10) for food in young adult (6 months) and aged (29 months) rats. THC dose-dependently decreased responding for food. Rimonabant alone had little or no effect on responding up to 1.0 mg/kg, but disrupted responding following a 3.2 mg/kg dose. Rimonabant (1.0 mg/kg) partially antagonized response disruption by THC. These effects were similar in young adult and aged rats. However, aging has been reported to change the neurobiology of cannabinoid CB1 receptors. To confirm our rats exhibited such differences, we assessed CB1 receptor binding sites and function in six subcortical (caudate, nucleus accumbens CA1, and CA2/CA3), and three cortical regions (medial prefrontal, temporal, entorhinal) in young adult (6 months) or aged (26 months) male Lewis rats using quantitative autoradiography. CB1 receptor binding sites were reduced in cortical, but not subcortical brain regions of aged rats. CB1 receptor function, at the level of receptor-G protein interaction, was not different in any region studied. Results indicate that down-regulation of CB1 receptor binding sites observed in cortical regions of aged rats was not accompanied by a commensurate decrease in CB1 receptor-stimulated [35S]GTPγS binding, suggesting a compensatory increase in receptor function in cortical areas. Together, our results provide additional evidence of age-related changes in central CB1 receptor populations. However, the functional compensation for decreased CB1 receptor binding may mitigate changes in behavioral effects of cannabinoids. With the rising use of cannabinoid-based therapeutics among aging populations, further evaluation of age-related changes in the cannabinoid system and the impact of these changes on effects of this class of drugs is warranted.
Collapse
Affiliation(s)
- Brett C. Ginsburg
- Department of Psychatry, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229
| | - Julie G. Hensler
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229,US Army Graduate Program in Anesthesia Nursing, US Army Medical Center of Excellence, 3490 Forage Rd., Suite 119, Joint Base San Antonio, Fort Sam Houston, Texas 78234-7585
| |
Collapse
|
19
|
Santos-Molina L, Herrerias A, Zawatsky CN, Gunduz-Cinar O, Cinar R, Iyer MR, Wood CM, Lin Y, Gao B, Kunos G, Godlewski G. Effects of a Peripherally Restricted Hybrid Inhibitor of CB1 Receptors and iNOS on Alcohol Drinking Behavior and Alcohol-Induced Endotoxemia. Molecules 2021; 26:5089. [PMID: 34443679 PMCID: PMC8399901 DOI: 10.3390/molecules26165089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/26/2022] Open
Abstract
Alcohol consumption is associated with gut dysbiosis, increased intestinal permeability, endotoxemia, and a cascade that leads to persistent systemic inflammation, alcoholic liver disease, and other ailments. Craving for alcohol and its consequences depends, among other things, on the endocannabinoid system. We have analyzed the relative role of central vs. peripheral cannabinoid CB1 receptors (CB1R) using a "two-bottle" as well as a "drinking in the dark" paradigm in mice. The globally acting CB1R antagonist rimonabant and the non-brain penetrant CB1R antagonist JD5037 inhibited voluntary alcohol intake upon systemic but not upon intracerebroventricular administration in doses that elicited anxiogenic-like behavior and blocked CB1R-induced hypothermia and catalepsy. The peripherally restricted hybrid CB1R antagonist/iNOS inhibitor S-MRI-1867 was also effective in reducing alcohol consumption after oral gavage, while its R enantiomer (CB1R inactive/iNOS inhibitor) was not. The two MRI-1867 enantiomers were equally effective in inhibiting an alcohol-induced increase in portal blood endotoxin concentration that was caused by increased gut permeability. We conclude that (i) activation of peripheral CB1R plays a dominant role in promoting alcohol intake and (ii) the iNOS inhibitory function of MRI-1867 helps in mitigating the alcohol-induced increase in endotoxemia.
Collapse
Affiliation(s)
- Luis Santos-Molina
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.S.-M.); (A.H.); (G.K.)
| | - Alexa Herrerias
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.S.-M.); (A.H.); (G.K.)
| | - Charles N. Zawatsky
- Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (C.N.Z.); (R.C.)
| | - Ozge Gunduz-Cinar
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Resat Cinar
- Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (C.N.Z.); (R.C.)
| | - Malliga R. Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (M.R.I.); (C.M.W.)
| | - Casey M. Wood
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (M.R.I.); (C.M.W.)
| | - Yuhong Lin
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (Y.L.); (B.G.)
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (Y.L.); (B.G.)
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.S.-M.); (A.H.); (G.K.)
| | - Grzegorz Godlewski
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.S.-M.); (A.H.); (G.K.)
| |
Collapse
|
20
|
Giménez-Gómez P, Ballestín R, Gil de Biedma-Elduayen L, Vidal R, Ferrer-Pérez C, Reguilón MD, O'Shea E, Miñarro J, Colado MI, Rodríguez-Arias M. Decreased kynurenine pathway potentiate resilience to social defeat effect on cocaine reward. Neuropharmacology 2021; 197:108753. [PMID: 34389399 DOI: 10.1016/j.neuropharm.2021.108753] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/26/2021] [Accepted: 08/08/2021] [Indexed: 02/08/2023]
Abstract
The kynurenine (KYN) pathway of tryptophan (TRP) degradation is activated by stress and inflammatory factors. It is now well established that social stress induces the activation of the immune system, with central inflammation and KYN metabolism being two of the main factors linking stress with depression. The aim of the present study was to evaluate the long-lasting changes in the KYN pathway induced by social defeat (SD) associated with the resilience or susceptibility to an increase in the conditioned rewarding effects of cocaine. Mice were exposed to repeated SD and 3 weeks later, a conditioned place preference (CPP) induced by a subthreshold dose of cocaine (1.5 mg/kg) was developed. KYN levels in plasma, cerebellum, hippocampus, striatum and limbic forebrain were studied at the end of the CPP procedure. Changes in the KYN pathway after exposure to pharmacological (oxytocin and indomethacin) and environmental interventions (environmental enrichment) were also evaluated. Our results showed that defeated susceptible (SD-S) mice had higher conditioning scores than resilient mice (SD-R). In addition, although KYN concentration was elevated in all defeated mice, SD-R mice showed smaller increases in KYN concentration in the cerebellum than SD-S mice. Oxytocin or Indomethacin treatment before SD normalized cocaine-induced CPP, although the increase in the KYN pathway was maintained. However, environmental enrichment before SD normalized cocaine-induced CPP and prevented the increase in the KYN pathway. The present study highlights the role of the KYN pathway and anti-inflammatory drugs acting on TRP metabolism as pharmacological targets to potentiate resilience to social stress effects.
Collapse
Affiliation(s)
- Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Raúl Ballestín
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain
| | - Leticia Gil de Biedma-Elduayen
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Rebeca Vidal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Carmen Ferrer-Pérez
- Departmento de Psicología and Sociología, Universidad de Zaragoza, C/ Ciudad Escolar s/n, 44003, Teruel, Spain
| | - Marina D Reguilón
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - José Miñarro
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - María Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Marta Rodríguez-Arias
- Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010, Valencia, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
21
|
Horikawa C, Otsuka R, Nishita Y, Tange C, Kato Y, Tanaka T, Rogi T, Shibata H, Ando F, Shimokata H. Interaction between cognitive leisure activity and long-chain polyunsaturated fatty acid intake on global cognitive decline in a Japanese longitudinal cohort study: National Institute for Longevity Sciences-Longitudinal Study of Aging. BMC Geriatr 2021; 21:443. [PMID: 34315440 PMCID: PMC8314584 DOI: 10.1186/s12877-021-02359-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/21/2021] [Indexed: 12/04/2022] Open
Abstract
Background There is a growing interest in the significance of adopting a variety of lifestyle habits for maintaining cognitive function among older adults. A lifestyle that is easy to modify, simple, and less burdensome for older people is ideal. We investigated the longitudinal association between global cognitive decline and cognitive leisure activities (CLAs) combined with long-chain polyunsaturated fatty acids (LCPUFAs) intake. Methods The National Institute for Longevity Sciences-Longitudinal Study of Aging (NILS-LSA) enrolled community-dwelling middle-aged and older men and women who were randomly selected from Obu-City and Higashiura Town, Aichi, Japan. Baseline data (2006–2008), including CLAs and dietary intake, were obtained from 517 participants (aged 60–84 years) with normal cognition. Global cognitive decline, defined as the Mini-Mental State Examination (MMSE) score ≤ 27, was assessed at baseline and four years later. Interaction between CLAs and LCPUFAs on cognitive decline was investigated using a multiple logistic analysis with adjustment for confounders. CLA engagement and LCPUFA intake were divided into high and low groups according to the frequency at which each participant engaged in the activity and the median intake level according to sex, respectively. Results A significant interaction was detected for the combination of CLA engagement and LCPUFA intake. Logistic regression coefficients revealed significant interactions when participants engaged in more than five CLA varieties. One of the CLAs, art appreciation, produced a significant main effect against cognitive decline and a significant interaction in combination with LCPUFA intake. The major LCPUFAs—docosahexaenoic acid and arachidonic acid—also exhibited a significant interaction. The combination of high LCPUFA intake and high art appreciation frequency yielded a lower adjusted odds ratio for cognitive decline than the combination of low LCPUFA and low art appreciation [0.25 (95 % confidence intervals, 0.11–0.56)]. Conclusions Preserving cognitive function might be associated with a combination of varied and high-frequency engagement in CLAs combined with high LCPUFA intake.
Collapse
Affiliation(s)
- Chika Horikawa
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, 474-8511, Obu-City, Aichi, Japan.,Institute for Health Care Science, Suntory Wellness Limited, 8-1-1 Seikadai, Seika-cho, Soraku- gun, 619-0284, Kyoto, Japan
| | - Rei Otsuka
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, 474-8511, Obu-City, Aichi, Japan.
| | - Yukiko Nishita
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, 474-8511, Obu-City, Aichi, Japan
| | - Chikako Tange
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, 474-8511, Obu-City, Aichi, Japan
| | - Yuki Kato
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, 474-8511, Obu-City, Aichi, Japan.,Faculty of Health and Medical Sciences, Aichi Shukutoku University, 2-9 Katahira, 480-1197, Nagakute- city, Aichi, Japan
| | - Takao Tanaka
- Institute for Health Care Science, Suntory Wellness Limited, 8-1-1 Seikadai, Seika-cho, Soraku- gun, 619-0284, Kyoto, Japan
| | - Tomohiro Rogi
- Institute for Health Care Science, Suntory Wellness Limited, 8-1-1 Seikadai, Seika-cho, Soraku- gun, 619-0284, Kyoto, Japan
| | - Hiroshi Shibata
- Institute for Health Care Science, Suntory Wellness Limited, 8-1-1 Seikadai, Seika-cho, Soraku- gun, 619-0284, Kyoto, Japan
| | - Fujiko Ando
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, 474-8511, Obu-City, Aichi, Japan.,Faculty of Health and Medical Sciences, Aichi Shukutoku University, 2-9 Katahira, 480-1197, Nagakute- city, Aichi, Japan
| | - Hiroshi Shimokata
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, 474-8511, Obu-City, Aichi, Japan.,Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, 57 Takenoyama, Iwasaki-cho, 470-0196, Nisshin-city, Aichi, Japan
| |
Collapse
|
22
|
Marszalek-Grabska M, Smaga I, Surowka P, Grochecki P, Slowik T, Filip M, Kotlinska JH. Memantine Prevents the WIN 55,212-2 Evoked Cross-Priming of Ethanol-Induced Conditioned Place Preference (CPP). Int J Mol Sci 2021; 22:ijms22157940. [PMID: 34360704 PMCID: PMC8348856 DOI: 10.3390/ijms22157940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022] Open
Abstract
The activation of the endocannabinoid system controls the release of many neurotransmitters involved in the brain reward pathways, including glutamate. Both endocannabinoid and glutamate systems are crucial for alcohol relapse. In the present study, we hypothesize that N-methyl-D-aspartate (NMDA) glutamate receptors regulate the ability of a priming dose of WIN 55,212-2 to cross-reinstate ethanol-induced conditioned place preference (CPP). To test this hypothesis, ethanol-induced (1.0 g/kg, 10% w/v, i.p.) CPP (unbiased method) was established using male adult Wistar rats. After CPP extinction, one group of animals received WIN 55,212-2 (1.0 and 2.0 mg/kg, i.p.), the cannabinoid receptor 1 (CB1) agonist, or ethanol, and the other group received memantine (3.0 or 10 mg/kg, i.p.), the NMDA antagonist and WIN 55,212-2 on the reinstatement day. Our results showed that a priming injection of WIN 55,212-2 (2.0 mg/kg, i.p.) reinstated (cross-reinstated) ethanol-induced CPP with similar efficacy to ethanol. Memantine (3.0 or 10 mg/kg, i.p.) pretreatment blocked this WIN 55,212-2 effect. Furthermore, our experiments indicated that ethanol withdrawal (7 days withdrawal after 10 days ethanol administration) down-regulated the CNR1 (encoding CB1), GRIN1/2A (encoding GluN1 and GluN2A subunit of the NMDA receptor) genes expression in the prefrontal cortex and dorsal striatum, but up-regulated these in the hippocampus, confirming the involvement of these receptors in ethanol rewarding effects. Thus, our results show that the endocannabinoid system is involved in the motivational properties of ethanol, and glutamate may control cannabinoid induced relapse into ethanol seeking behavior.
Collapse
Affiliation(s)
- Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (I.S.); (M.F.)
| | - Paulina Surowka
- Affective Cognitive Neuroscience Laboratory, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland;
| | - Pawel Grochecki
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Tymoteusz Slowik
- Experimental Medicine Center, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Malgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (I.S.); (M.F.)
| | - Jolanta H. Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 20-093 Lublin, Poland;
- Correspondence:
| |
Collapse
|
23
|
Dulman RS, Zhang H, Banerjee R, Krishnan HR, Dong B, Hungund BL, Vinod KY, Pandey SC. CB1 receptor neutral antagonist treatment epigenetically increases neuropeptide Y expression and decreases alcohol drinking. Neuropharmacology 2021; 195:108623. [PMID: 34048869 DOI: 10.1016/j.neuropharm.2021.108623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/22/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Alcohol consumption is mediated by several important neuromodulatory systems, including the endocannabinoid and neuropeptide Y (NPY) systems in the limbic brain circuitry. However, molecular mechanisms through which cannabinoid-1 (CB1) receptors regulate alcohol consumption are still unclear. Here, we investigated the role of the CB1 receptor-mediated downstream regulation of NPY via epigenetic mechanisms in the amygdala. Alcohol drinking behavior was measured in adult male C57BL/6J mice treated with a CB1 receptor neutral antagonist AM4113 using a two-bottle choice paradigm while anxiety-like behavior was assessed in the light-dark box (LDB) test. The CB1 receptor-mediated changes in the protein levels of phosphorylated cAMP-responsive element binding protein (pCREB), CREB binding protein (CBP), H3K9ac, H3K14ac and NPY, and the mRNA levels of Creb1, Cbp, and Npy were measured in amygdaloid brain structures. Npy-specific changes in the levels of acetylated histone (H3K9/14ac) and CBP in the amygdala were also measured. We found that the pharmacological blockade of CB1 receptors with AM4113 reduced alcohol consumption and, in an ethanol-naïve cohort, reduced anxiety-like behavior in the LDB test. Treatment with AM4113 also increased the mRNA levels of Creb1 and Cbp in the amygdala as well as the protein levels of pCREB, CBP, H3K9ac and H3K14ac in the central and medial nucleus of amygdala, but not in the basolateral amygdala. Additionally, AM4113 treatment increased occupancy of CBP and H3K9/14ac at the Npy gene promoter, leading to an increase in both mRNA and protein levels of NPY in the amygdala. These novel findings suggest that CB1 receptor-mediated CREB signaling plays an important role in the modulation of NPY function through an epigenetic mechanism and further support the potential use of CB1 receptor neutral antagonists for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Russell S Dulman
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| | - Ritabrata Banerjee
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Harish R Krishnan
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| | - Bin Dong
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Basalingappa L Hungund
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA; New York State Psychiatric Institute, New York, NY, 10032, USA
| | - K Yaragudri Vinod
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA; Emotional Brain Institute, Orangeburg, NY, 10962, USA; Department of Child and Adolescent Psychiatry, New York School of Medicine, New York, NY, 10016, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
24
|
Manca C, Carta G, Murru E, Abolghasemi A, Ansar H, Errigo A, Cani PD, Banni S, Pes GM. Circulating fatty acids and endocannabinoidome-related mediator profiles associated to human longevity. GeroScience 2021; 43:1783-1798. [PMID: 33650014 PMCID: PMC8492808 DOI: 10.1007/s11357-021-00342-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
To evaluate whether a peculiar plasma profile of fatty acids and endocannabinoidome (eCBome)-related mediators may be associated to longevity, we assessed them in octogenarians (Old; n=42) living in the east-central mountain area of Sardinia, a High-Longevity Zone (HLZ), compared to sexagenarian (Young; n=21) subjects from the same area, and to Olds (n=22) from the Northern Sardinia indicated as Lower-Longevity Zone (LLZ). We found significant increases in conjugated linoleic acid (CLA) and heptadecanoic acid (17:0) levels in Old-HLZ with respect to younger subjects and Old-LLZ subjects. Young-HLZ subjects exhibited higher circulating levels of pentadecanoic acid (15:0) and retinol. Palmitoleic acid (POA) was elevated in both Young and Old subjects from the HLZ. eCBome profile showed a significantly increased plasma level of the two endocannabinoids, N-arachidonoyl-ethanolamine (AEA) and 2-arachidonoyl-glycerol (2-AG) in Old-HLZ subjects compared to Young-HLZ and Old-LLZ respectively. In addition, we found increased N-oleoyl-ethanolamine (OEA), 2-linoleoyl-glycerol (2-LG) and 2-oleoyl-glycerol (2-OG) levels in Old-HLZ group with respect to Young-HLZ (as for OEA an d 2-LG) and both the Old-LLZ and Young-HLZ for 2-OG. The endogenous metabolite of docosahexaenoic acid (DHA), N-docosahexaenoyl-ethanolamine (DHEA) was significantly increased in Old-HLZ subjects. In conclusion, our results suggest that in the HLZ area, Young and Old subjects exhibited a favourable, albeit distinctive, fatty acids and eCBome profile that may be indicative of a metabolic pattern potentially protective from adverse chronic conditions. These factors could point to a suitable physiological metabolic pattern that may counteract the adverse stimuli leading to age-related disorders such as neurodegenerative and metabolic diseases.
Collapse
Affiliation(s)
- Claudia Manca
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Monserrato, CA, Italy
| | - Gianfranca Carta
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Monserrato, CA, Italy
| | - Elisabetta Murru
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Monserrato, CA, Italy
| | - Armita Abolghasemi
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Monserrato, CA, Italy
| | - Hastimansooreh Ansar
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Monserrato, CA, Italy
| | - Alessandra Errigo
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.,WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Brussels, Belgium
| | - Sebastiano Banni
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Monserrato, CA, Italy.
| | - Giovanni Mario Pes
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy.,Sardinia Longevity Blue Zone Observatory, Ogliastra, Italy
| |
Collapse
|
25
|
Oppong-Damoah A, Gannon BM, Murnane KS. The Endocannabinoid System and Alcohol Dependence: Will Cannabinoid Receptor 2 Agonism be More Fruitful than Cannabinoid Receptor 1 Antagonism? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2021; 21:3-13. [PMID: 33573565 DOI: 10.2174/1871527320666210211115007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 11/22/2022]
Abstract
Alcohol-use disorder (AUD) remains a major public health concern. In recent years, there has been a heightened interest in components of the endocannabinoid system for the treatment of AUD. Cannabinoid type 1 (CB1) receptors have been shown to modulate the rewarding effects of alcohol, reduce the abuse-related effects of alcohol, improve cognition, exhibit anti-inflammatory, and neuroprotective effects, which are all favorable properties of potential therapeutic candidates for the treatment of AUD. However, CB1 agonists have not been investigated for the treatment of AUD because they stimulate the motivational properties of alcohol, increase alcohol intake, and have the tendency to be abused. Preclinical data suggest significant potential for the use of CB1 antagonists to treat AUD; however, a clinical phase I/II trial with SR14716A (rimonabant), a CB1 receptor antagonist/inverse agonist showed that it produced serious neuropsychiatric adverse events such as anxiety, depression, and even suicidal ideation. This has redirected the field to focus on alternative components of the endocannabinoid system, including cannabinoid type 2 (CB2) receptor agonists as a potential therapeutic target for AUD. CB2 receptor agonists are of particular interest because they can modulate the reward pathway, reduce abuse-related effects of alcohol, reverse neuroinflammation, improve cognition, and exhibit anti-inflammatory and neuroprotective effects, without exhibiting the psychiatric side effects seen with CB1 antagonists. Accordingly, this article presents an overview of the studies reported in the literature that have investigated CB2 receptor agonists with regards to AUD and provides commentary as to whether this receptor is a worthy target for continued investigation.
Collapse
Affiliation(s)
- Aboagyewaah Oppong-Damoah
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center,United States
| | - Brenda Marie Gannon
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center,United States
| | - Kevin Sean Murnane
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center,United States
| |
Collapse
|
26
|
Morales-Puerto N, Giménez-Gómez P, Pérez-Hernández M, Abuin-Martínez C, Gil de Biedma-Elduayen L, Vidal R, Gutiérrez-López MD, O'Shea E, Colado MI. Addiction and the kynurenine pathway: A new dancing couple? Pharmacol Ther 2021; 223:107807. [PMID: 33476641 DOI: 10.1016/j.pharmthera.2021.107807] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
Drug use poses a serious threat to health systems throughout the world and the number of consumers rises relentlessly every year. The kynurenine pathway, main pathway of tryptophan degradation, has drawn interest in this field due to its relationship with addictive behaviour. Recently it has been confirmed that modulation of kynurenine metabolism at certain stages of the pathway can reduce, prevent or abolish drug seeking-like behaviours in studies with several different drugs. In this review, we present an up-to-date summary of the evidences of a relationship between drug use and the kynurenine pathway, both the alterations of the pathway due to drug use as well as modulation of the pathway as a potential approach to treat drug addiction. The review discusses ethanol, nicotine, cannabis, amphetamines, cocaine and opioids and new prospects in the drug research field are proposed.
Collapse
Affiliation(s)
- Nuria Morales-Puerto
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Cristina Abuin-Martínez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Leticia Gil de Biedma-Elduayen
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Rebeca Vidal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María Dolores Gutiérrez-López
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.
| | - María Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
27
|
Durieux LJA, Gilissen SRJ, Arckens L. Endocannabinoids and cortical plasticity: CB1R as a possible regulator of the excitation/inhibition balance in health and disease. Eur J Neurosci 2021; 55:971-988. [PMID: 33427341 DOI: 10.1111/ejn.15110] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/27/2022]
Abstract
The endocannabinoid system has been linked to neurological disorders in which the excitation inhibition (E/I) balance in the neocortex is dysregulated, such as schizophrenia. The main endocannabinoid receptor type 1 of the central nervous system-CB1R-is expressed on different cell types, that when activated, modulate the cortical E/I balance. Here we review how CB1R signalling contributes to phases of heightened plasticity of the neocortex. We review the major role of the CB1R in cortical plasticity throughout life, including the early life sensory critical periods, the later maturation phase of the association cortex in adolescence, and the adult phase of sensory deprivation-induced cortical plasticity. Endocannabinoid-mediated long-term potentiation and depression of synapse strength fine-tune the E/I balance in visual, somatosensory and association areas. We emphasize how a distinct set of key endocannabinoid-regulated elements such as GABA and glutamate release, basket parvalbumin interneurons, somatostatin interneurons and astrocytes, are essential for normal cortical plasticity and dysregulated in schizophrenia. Even though a lot of data has been gathered, mechanistic knowledge about the exact CB1R-based modulation of excitation and/or inhibition is still lacking depending on cortical area and maturation phase in life. We emphasize the importance of creating such detailed knowledge for a better comprehension of what underlies the dysregulation of the neocortex in schizophrenic patients in adulthood. We propose that taking age, brain area and cell type into consideration when modulating the cortical E/I imbalance via cannabinoid-based pharmacology may pave the way for better patient care.
Collapse
Affiliation(s)
- Lucas J A Durieux
- KU Leuven, Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| | - Sara R J Gilissen
- KU Leuven, Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| | - Lutgarde Arckens
- KU Leuven, Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
28
|
Buch C, Muller T, Leemput J, Passilly-Degrace P, Ortega-Deballon P, Pais de Barros JP, Vergès B, Jourdan T, Demizieux L, Degrace P. Endocannabinoids Produced by White Adipose Tissue Modulate Lipolysis in Lean but Not in Obese Rodent and Human. Front Endocrinol (Lausanne) 2021; 12:716431. [PMID: 34434170 PMCID: PMC8382141 DOI: 10.3389/fendo.2021.716431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
White adipose tissue (WAT) possesses the endocannabinoid system (ECS) machinery and produces the two major endocannabinoids (ECs), arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG). Accumulating evidence indicates that WAT cannabinoid 1 receptors (CB1R) are involved in the regulation of fat storage, tissue remodeling and secretory functions but their role in controlling lipid mobilization is unclear. In the present study, we used different strategies to acutely increase ECS activity in WAT and tested the consequences on glycerol production as a marker of lipolysis. Treating lean mice or rat WAT explants with JLZ195, which inhibits ECs degrading enzymes, induced an increase in 2-AG tissue contents that was associated with a CB1R-dependent decrease in lipolysis. Direct treatment of rat WAT explants with AEA also inhibited glycerol production while mechanistic studies revealed it could result from the stimulation of Akt-signaling pathway. Interestingly, AEA treatment decreased lipolysis both in visceral and subcutaneous WAT collected on lean subjects suggesting that ECS also reduces fat store mobilization in Human. In obese mice, WAT content and secretion rate of ECs were higher than in control while glycerol production was reduced suggesting that over-produced ECs may inhibit lipolysis activating local CB1R. Strikingly, our data also reveal that acute CB1R blockade with Rimonabant did not modify lipolysis in vitro in obese mice and human explants nor in vivo in obese mice. Taken together, these data provide physiological evidence that activation of ECS in WAT, by limiting fat mobilization, may participate in the progressive tissue remodeling that could finally lead to organ dysfunction. The present findings also indicate that acute CB1R blockade is inefficient in regulating lipolysis in obese WAT and raise the possibility of an alteration of CB1R signaling in conditions of obesity.
Collapse
Affiliation(s)
- Chloé Buch
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Tania Muller
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Julia Leemput
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Patricia Passilly-Degrace
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pablo Ortega-Deballon
- Department of Digestive, Thoracic and Surgical Oncology, University Hospital, Dijon, France
| | | | - Bruno Vergès
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
- Department of Endocrinology-Diabetology, University Hospital, Dijon, France
| | - Tony Jourdan
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Laurent Demizieux
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pascal Degrace
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
- *Correspondence: Pascal Degrace,
| |
Collapse
|
29
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
30
|
Bobrich M, Schwarz R, Ramer R, Borchert P, Hinz B. A simple LC-MS/MS method for the simultaneous quantification of endocannabinoids in biological samples. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1161:122371. [DOI: 10.1016/j.jchromb.2020.122371] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/30/2020] [Accepted: 09/02/2020] [Indexed: 01/15/2023]
|
31
|
Pavon FJ, Polis I, Stouffer DG, Roberto M, Martin-Fardon R, Rodriguez de Fonseca F, Parsons LH, Serrano A. COX-2 Inhibition Antagonizes Intra-Accumbens 2-Arachidonoylglycerol-Mediated Reduction in Ethanol Self-Administration in Rats. Alcohol Clin Exp Res 2020; 44:2158-2165. [PMID: 32944989 DOI: 10.1111/acer.14456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Ethanol (EtOH) self-administration is particularly sensitive to the modulation of CB1 signaling in the nucleus accumbens (NAc) shell, and EtOH consumption increases extracellular levels of the endogenous cannabinoid CB1 receptor agonist 2-arachidonoyl glycerol (2-AG) in this brain region. Stimulation of CB1 receptor with agonists increases EtOH consumption, suggesting that EtOH-induced increases in 2-AG might sustain motivation for EtOH intake. METHODS In order to further explore this hypothesis, we analyzed the alterations in operant EtOH self-administration induced by intra-NAc shell infusions of 2-AG itself, the CB1 inverse agonist SR141716A, the 2-AG clearance inhibitor URB602, anandamide, and the cyclooxygenase-2 (COX-2) inhibitor nimesulide. RESULTS Surprisingly, self-administration of 10% EtOH was dose-dependently reduced by either intra-NAc shell SR141716A or 2-AG infusions. Similar effects were found by intra-NAc shell infusions of URB602, suggesting again a role for accumbal 2-AG on the modulation of EtOH intake. Intra-NAc shell anandamide did not alter EtOH self-administration, pointing to a specific role for 2-AG in the modulation of EtOH self-administration. Finally, the inhibitory effect of intra-NAc shell 2-AG on EtOH intake was significantly reversed by pretreatment with nimesulide, suggesting that oxidative metabolites of 2-AG might mediate these inhibitory effects on operant self-administration. CONCLUSIONS We propose that 2-AG signaling in the NAc exerts an inhibitory influence on EtOH consumption through a non-CB1 receptor mechanism involving the COX-2 pathway.
Collapse
Affiliation(s)
- Francisco J Pavon
- From the, Department of Neuroscience, (FJP, IP, DGS, LHP, AS), The Scripps Research Institute, La Jolla, California, USA.,Instituto de Investigación Biomédica de Málaga, (IBIMA) (FJP, FRF, AS), Hospital Regional Universitario de Málaga, Unidad de Gestión Clínica de Salud Mental, Malaga, Spain
| | - Ilham Polis
- From the, Department of Neuroscience, (FJP, IP, DGS, LHP, AS), The Scripps Research Institute, La Jolla, California, USA
| | - David G Stouffer
- From the, Department of Neuroscience, (FJP, IP, DGS, LHP, AS), The Scripps Research Institute, La Jolla, California, USA
| | - Marisa Roberto
- Department of Molecular Medicine, (MR, RM-F), The Scripps Research Institute, La Jolla, California, USA
| | - Rémi Martin-Fardon
- Department of Molecular Medicine, (MR, RM-F), The Scripps Research Institute, La Jolla, California, USA
| | - Fernando Rodriguez de Fonseca
- Instituto de Investigación Biomédica de Málaga, (IBIMA) (FJP, FRF, AS), Hospital Regional Universitario de Málaga, Unidad de Gestión Clínica de Salud Mental, Malaga, Spain
| | - Loren H Parsons
- From the, Department of Neuroscience, (FJP, IP, DGS, LHP, AS), The Scripps Research Institute, La Jolla, California, USA
| | - Antonia Serrano
- From the, Department of Neuroscience, (FJP, IP, DGS, LHP, AS), The Scripps Research Institute, La Jolla, California, USA.,Instituto de Investigación Biomédica de Málaga, (IBIMA) (FJP, FRF, AS), Hospital Regional Universitario de Málaga, Unidad de Gestión Clínica de Salud Mental, Malaga, Spain
| |
Collapse
|
32
|
Suárez J, Khom S, Alén F, Natividad LA, Varodayan FP, Patel RR, Kirson D, Arco R, Ballesta A, Bajo M, Rubio L, Martin-Fardon R, de Fonseca FR, Roberto M. Cessation of fluoxetine treatment increases alcohol seeking during relapse and dysregulates endocannabinoid and glutamatergic signaling in the central amygdala. Addict Biol 2020; 25:e12813. [PMID: 31339221 PMCID: PMC8050940 DOI: 10.1111/adb.12813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/17/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023]
Abstract
Administration of selective serotonin reuptake inhibitors (SSRIs), typically used as antidepressants, induces long-lasting behavioral changes associated with alcohol use disorder (AUD). However, the contribution of SSRI (fluoxetine)-induced alterations in neurobiological processes underlying alcohol relapse such as endocannabinoid and glutamate signaling in the central amygdala (CeA) remains largely unknown. We utilized an integrative approach to study the effects of repeated fluoxetine administration during abstinence on ethanol drinking. Gene expression and biochemical and electrophysiological studies explored the hypothesis that dysregulation in glutamatergic and endocannabinoid mechanisms in the CeA underlie the susceptibility to alcohol relapse. Cessation of daily treatment with fluoxetine (10 mg/kg) during abstinence resulted in a marked increase in ethanol seeking during re-exposure periods. The increase in ethanol self-administration was associated with (a) reductions in levels of the endocannabinoids N-arachidonoylethanolomine and 2-arachidonoylglycerol in the CeA, (b) increased amygdalar gene expression of cannabinoid type-1 receptor (CB1), N-acyl phosphatidylethanolamine phospholipase D (Nape-pld), fatty acid amid hydrolase (Faah), (c) decreased amygdalar gene expression of ionotropic AMPA (GluA2 and GluA4) and metabotropic (mGlu3) glutamate receptors, and (d) increased glutamatergic receptor function. Overall, our data suggest that the administration of the antidepressant fluoxetine during abstinence dysregulates endocannabinoid signaling and glutamatergic receptor function in the amygdala, facts that likely facilitate alcohol drinking behavior during relapse.
Collapse
Affiliation(s)
- Juan Suárez
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
- Fulbright Visiting Scholar Program, Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Sophia Khom
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Francisco Alén
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
- Department of Psychobiology. Universidad Complutense de Madrid, Madrid, Spain
| | - Luis A. Natividad
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Florence P. Varodayan
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Reesha R. Patel
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Dean Kirson
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Rocío Arco
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
| | - Antonio Ballesta
- Department of Psychobiology. Universidad Complutense de Madrid, Madrid, Spain
| | - Michal Bajo
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Leticia Rubio
- Department of Anatomy and Forensic and Legal Medicine. Universidad de Málaga, Málaga, Spain
| | - Rémi Martin-Fardon
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
- Department of Psychobiology. Universidad Complutense de Madrid, Madrid, Spain
| | - Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| |
Collapse
|
33
|
Millie LA, Boehm SL, Grahame NJ. Attentional set shifting in HAP3, LAP3, and cHAP mice is unaffected by either genetic differences in alcohol preference or an alcohol drinking history. Exp Clin Psychopharmacol 2020; 28:379-387. [PMID: 32150428 PMCID: PMC7390659 DOI: 10.1037/pha0000359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Alcohol consumption may precede, or result from, behavioral inflexibility and contribute to individuals' difficulties ceasing drinking. Attentional set shifting tasks are an animal analog to a human behavioral flexibility task requiring recognition of a previous strategy as inappropriate, and the formation and maintenance of a novel strategy (Floresco, Block, & Tse, 2008). Abstinent individuals with alcohol use disorder, nonalcoholic individuals with a family history of alcoholism, and mice exposed to chronic-intermittent alcohol vapor show impaired behavioral flexibility (Gierski et al., 2013; Hu, Morris, Carrasco, & Kroener, 2015; Oscar-Berman et al., 2009). Behavioral flexibility deficits can be linked to frontal cortical regions connected to the striatum (Ragozzino, 2007), and alterations to the endocannabinoid system, implicated in drug seeking and consumption (Economidou et al., 2006; Serrano & Parsons, 2011), may affect these behaviors. Alcohol-preferring and nonpreferring rodents exhibit differences in CB1 receptor expression (CB1R; Hansson et al., 2007; Hungund & Basavarajappa, 2000), but whether dorsal striatal CB1Rs are important for other alcohol-related behaviors such as attentional set shifting tasks remains unclear. This study assesses whether selectively bred high (HAP) versus low alcohol-preferring mice differ in an operant attentional set shifting task or CB1R levels in the dorsal striatum and whether a history of voluntary alcohol consumption in crossed HAP mice exacerbates inflexibility. Contrary to our hypothesis, neither genetic differences in alcohol preference nor drinking affected set shifting. However, high alcohol-preferring mice-3 mice showed reduced levels of dorsal striatal CB1R compared with low alcohol-preferring-3 mice, suggesting that genetic differences in alcohol consumption may be mediated in part by striatal CB1R. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
Affiliation(s)
- Lauren A Millie
- Department of Psychology, Indiana University-Purdue University Indianapolis
| | - Stephen L Boehm
- Department of Psychology, Indiana University-Purdue University Indianapolis
| | - Nicholas J Grahame
- Department of Psychology, Indiana University-Purdue University Indianapolis
| |
Collapse
|
34
|
Li R, Huang Z, Luo J, Luo H, Wang W. Downregulation of the CB1-Mediated Endocannabinoid Signaling Underlies D-Galactose-Induced Memory Impairment. Front Mol Neurosci 2020; 13:130. [PMID: 32848596 PMCID: PMC7399637 DOI: 10.3389/fnmol.2020.00130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/26/2020] [Indexed: 12/17/2022] Open
Abstract
Imbalance in redox homeostasis is a major cause of age-related cognitive impairment. The endocannabinoid system (ECS) is a key player in regulating synaptic transmission, plasticity and memory. Increasing evidence indicates an important interplay between the two systems. However, how excessive oxidative stress could alter ECS and that, in turn, impairs its modulatory role in synaptic plasticity and cognitive function remains elusive. In the present study, we examined this causal link in D-galactose-induced oxidative rats. First, the reactive oxygen species generating enzymes, especially nitric oxide synthase (NOS), indeed show an elevated expression in D-galactose-treated rats, and this was correlated to an impaired hippocampal long-term potentiation (LTP) and spatial memory loss in animal behavioral tests. Second, the cannabinoid receptor type I (CB1)-mediated signaling is known to regulate synaptic plasticity. We show that a decrease in CB1 and increase in degradation enzymes for CB1 ligand endocannabinoid anandamide all occurred to D-galactose-treated rats. Surprisingly, application of low-dose anandamide, known to reduce LTP under physiological condition, now acted to enhance LTP in D-galactose-treated rats, most likely resulted from the inhibition of GABAergic synapses. Furthermore, this reversal behavior of CB1-signaling could be fully simulated by a NOS inhibitor, diphenyleneiodonium. These observations suggest that interaction between redox dysfunction and ECS should contribute significantly to the impaired synaptic plasticity and memory loss in D-galactose-treated rats. Therefore, therapies focusing on the balance of these two systems may shed lights on the treatment of age-related cognitive impairment in the future.
Collapse
Affiliation(s)
- Ranran Li
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Huang
- Department of Anesthesia, Wuhan Fourth Hospital and Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Luo
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Luo
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Hauser SR, Katner SN, Waeiss RA, Truitt WA, Bell RL, McBride WJ, Rodd ZA. Selective breeding for high alcohol preference is associated with increased sensitivity to cannabinoid reward within the nucleus accumbens shell. Pharmacol Biochem Behav 2020; 197:173002. [PMID: 32710885 DOI: 10.1016/j.pbb.2020.173002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 11/30/2022]
Abstract
RATIONALE The rate of cannabinoid intake by those with alcohol use disorder (AUD) exceeds that of the general public. The high prevalence of co-abuse of alcohol and cannabis has been postulated to be predicated upon both a common predisposing genetic factor and the interaction of the drugs within the organism. The current experiments examined the effects of cannabinoids in an animal model of AUD. OBJECTIVES The present study assessed the reinforcing properties of a cannabinoid receptor 1 (CB1) agonist self-administered directly into the nucleus accumbens shell (AcbSh) in female Wistar and alcohol-preferring (P) rats. METHODS Following guide cannulae surgery aimed at AcbSh, subjects were placed in an operant box equipped with an 'active lever' (fixed ratio 1; FR1) that caused the delivery of the infusate and an 'inactive lever' that did not. Subjects were arbitrarily assigned to one of seven groups that self-administered either artificial cerebrospinal fluid (aCSF), or 3.125, 6.25, 12.5, or 25 pmol/100 nl of O-1057, a water-soluble CB1 agonist, dissolved in aCSF. The first four sessions of acquisition are followed by aCSF only infusates in sessions 5 and 6 during extinction, and finally the acquisition dose of infusate during session 7 as reinstatement. RESULTS The CB1 agonist was self-administered directly into the AcbSh. P rats self-administered the CB1 agonist at lower concentrations and at higher rates compared to Wistar rats. CONCLUSIONS Overall, the data indicate selective breeding for high alcohol preference has produced rats divergent in response to cannabinoids within the brain reward pathway. The data support the hypothesis that there can be common genetic factors influencing drug addiction.
Collapse
Affiliation(s)
- Sheketha R Hauser
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Simon N Katner
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Robert A Waeiss
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William A Truitt
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard L Bell
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William J McBride
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zachary A Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
36
|
Hodges EL, Marshall JP, Ashpole NM. Age-dependent hormesis-like effects of the synthetic cannabinoid CP55940 in C57BL/6 mice. NPJ Aging Mech Dis 2020; 6:7. [PMID: 32655880 PMCID: PMC7338393 DOI: 10.1038/s41514-020-0045-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 06/16/2020] [Indexed: 11/13/2022] Open
Abstract
Use of cannabis and cannabinoid-containing substances is increasing among geriatric patients, despite relatively sparse preclinical evidence in aged models. To better understand the effects of exogenous cannabinoids on aging male and female rodents, we compared the age- and dose-dependent physiological and behavioral effects of the synthetic cannabinoid CP55940 in young-adult and aged C57BL/6 mice. Locomotion, body temperature, thermal nociception, and fecal output were measured following CP55940 administration. Our findings indicate that CP55940 is more potent and efficacious in older mice, evidenced by exaggerated antinociception and locomotor inhibition when compared to younger adult mice. In addition, we report that low doses of CP55940 paradoxically stimulate locomotion in young-adult (4 m) mice; however, this hormesis-like response is not as evident in aged animals (21-24 m). These bidirectional effects appear to be mediated via the endocannabinoid CB1 and CB2 receptors.
Collapse
Affiliation(s)
- Erik L. Hodges
- Department of BioMolecular Sciences, Pharmacology Division, University of Mississippi School of Pharmacy, University, Oxford, MS USA
| | - Jessica P. Marshall
- Department of BioMolecular Sciences, Pharmacology Division, University of Mississippi School of Pharmacy, University, Oxford, MS USA
| | - Nicole M. Ashpole
- Department of BioMolecular Sciences, Pharmacology Division, University of Mississippi School of Pharmacy, University, Oxford, MS USA
- Research Institute of Pharmaceutical Sciences, University of Mississippi School of Pharmacy, University, Oxford, MS USA
| |
Collapse
|
37
|
Cannabinoid receptor CNR1 expression and DNA methylation in human prefrontal cortex, hippocampus and caudate in brain development and schizophrenia. Transl Psychiatry 2020; 10:158. [PMID: 32433545 PMCID: PMC7237456 DOI: 10.1038/s41398-020-0832-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 12/22/2022] Open
Abstract
Beyond being one the most widely used psychoactive drugs in the world, cannabis has been identified as an environmental risk factor for psychosis. Though the relationship between cannabis use and psychiatric disorders remains controversial, consistent association between early adolescent cannabis use and the subsequent risk of psychosis suggested adolescence may be a particularly vulnerable period. Previous findings on gene by environment interactions indicated that cannabis use may only increase the risk for psychosis in the subjects who have a specific genetic vulnerability. The type 1 cannabinoid receptor (CB1), encoded by the CNR1 gene, is a key component of the endocannabinoid system. As the primary endocannabinoid receptor in the brain, CB1 is the main molecular target of the endocannabinoid ligand, as well as tetrahydrocannabinol (THC), the principal psychoactive ingredient of cannabis. In this study, we have examined mRNA expression and DNA methylation of CNR1 in human prefrontal cortex (PFC), hippocampus, and caudate samples. The expression of CNR1 is higher in fetal PFC and hippocampus, then drops down dramatically after birth. The lifespan trajectory of CNR1 expression in the DLPFC differentially correlated with age by allelic variation at rs4680, a functional polymorphism in the COMT gene. Compared with COMT methionine158 carriers, Caucasian carriers of the COMT valine158 allele have a stronger negative correlation between the expression of CNR1 in DLPFC and age. In contrast, the methylation level of cg02498983, which is negatively correlated with the expression of CNR1 in PFC, showed the strongest positive correlation with age in PFC of Caucasian carriers of COMT valine158. Additionally, we have observed decreased mRNA expression of CNR1 in the DLPFC of patients with schizophrenia. Further analysis revealed a positive eQTL SNP, rs806368, which predicted the expression of a novel transcript of CNR1 in human DLPFC, hippocampus and caudate. This SNP has been associated with addiction and other psychiatric disorders. THC or ethanol are each significantly associated with dysregulated expression of CNR1 in the PFC of patients with affective disorder, and the expression of CNR1 is significantly upregulated in the PFC of schizophrenia patients who completed suicide. Our results support previous studies that have implicated the endocannabinoid system in the pathology of schizophrenia and provided additional insight into the mechanism of increasing risk for schizophrenia in the adolescent cannabis users.
Collapse
|
38
|
Kataoka K, Bilkei-Gorzo A, Nozaki C, Togo A, Nakamura K, Ohta K, Zimmer A, Asahi T. Age-dependent Alteration in Mitochondrial Dynamics and Autophagy in Hippocampal Neuron of Cannabinoid CB1 Receptor-deficient Mice. Brain Res Bull 2020; 160:40-49. [PMID: 32294520 DOI: 10.1016/j.brainresbull.2020.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Endocannabinoid system activity contributes to the homeostatic defense against aging and thus may counteract the progression of brain aging. The cannabinoid type 1 (CB1) receptor activity declines with aging in the brain, which impairs neuronal network integrity and cognitive functions. However, the underlying mechanisms that link CB1 activity and memory decline remain unknown. Mitochondrial activity profoundly influences neuronal function, and age-dependent mitochondrial activity change is one of the known hallmarks of brain aging. As CB1 receptor is expressed on mitochondria and may regulate neuronal energy metabolism in hippocampus, we hypothesized that CB1 receptors might influence mitochondria in hippocampal neurons. Here, we found that CB1 receptor significantly affected mitochondrial autophagy (mitophagy) and morphology in an age-dependent manner. Serine 65-phosphorylated ubiquitin, a key marker for mitophagy, was reduced in adult CB1-deficient mice (CB1-KO) compared to those in wild type controls, particularly in CA1 pyramidal cell layer. Transmission electron microscopy (TEM) analysis showed reduced mitophagy-like events in hippocampus of adult CB1-KO. TEM analysis also showed that mitochondrial morphology in adult CB1-KO mice was altered shown by an increase in thin and elongated mitochondria in hippocampal neurons. 3D reconstruction of mitochondrial morphology after scanning electron microscopy additionally revealed an enhanced density of interconnected mitochondria. Altogether, these findings suggest that reduced CB1 signaling in CB1-KO mice leads to reduced mitophagy and abnormal mitochondrial morphology in hippocampal neurons during aging. These mitochondrial changes might be due to the impairments in mitochondrial quality control system, which links age-related decline in CB1 activity and impaired memory.
Collapse
Affiliation(s)
- Kosuke Kataoka
- Faculty of Science and Engineering, Waseda University, 169-8555, Shinjuku, Tokyo, Japan; Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
| | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Chihiro Nozaki
- Faculty of Science and Engineering, Waseda University, 169-8555, Shinjuku, Tokyo, Japan; Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, 830-0011, Kurume, Fukuoka, Japan
| | - Keiichiro Nakamura
- Division Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, 830-0011, Kurume, Fukuoka, Japan
| | - Keisuke Ohta
- Advanced Imaging Research Center, Kurume University School of Medicine, 830-0011, Kurume, Fukuoka, Japan
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Toru Asahi
- Faculty of Science and Engineering, Waseda University, 169-8555, Shinjuku, Tokyo, Japan.
| |
Collapse
|
39
|
Kunos G. Interactions Between Alcohol and the Endocannabinoid System. Alcohol Clin Exp Res 2020; 44:790-805. [PMID: 32056226 DOI: 10.1111/acer.14306] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022]
Abstract
Endocannabinoids are lipid mediators that interact with the same cannabinoid receptors that recognize Δ9 -tetrahydrocannabinol (THC), the psychoactive constituent of marijuana, to induce similar effects in the brain and periphery. Alcohol and THC are both addictive substances whose acute use elicits rewarding effects that can lead to chronic and compulsive use via engaging similar signaling pathways in the brain. In the liver, both alcohol and endocannabinoids activate lipogenic gene expression leading to fatty liver disease. This review focuses on evidence accumulated over the last 2 decades to indicate that both the addictive neural effects of ethanol and its organ toxic effects in the liver and elsewhere are mediated, to a large extent, by endocannabinoids signaling via cannabinoid-1 receptors (CB1 R). The therapeutic potential of CB1 R blockade globally or in peripheral tissues only is also discussed.
Collapse
Affiliation(s)
- George Kunos
- From the, Division of Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
40
|
Abstract
Given the aging Baby Boomer generation, changes in cannabis legislation, and the growing acknowledgment of cannabis for its therapeutic potential, it is predicted that cannabis use in the older population will escalate. It is, therefore, important to determine the interaction between the effects of cannabis and aging. The aim of this report is to describe the link between cannabis use and the aging brain. Our review of the literature found few and inconsistent empirical studies that directly address the impact of cannabis use on the aging brain. However, research focused on long-term cannabis use points toward cumulative effects on multimodal systems in the brain that are similarly affected during aging. Specifically, the effects of cannabis and aging converge on overlapping networks in the endocannabinoid, opioid, and dopamine systems that may affect functional decline particularly in the hippocampus and prefrontal cortex, which are critical areas for memory and executive functioning. To conclude, despite the limited current knowledge on the potential interactive effects between cannabis and aging, evidence from the literature suggests that cannabis and aging effects are concurrently present across several neurotransmitter systems. There is a great need for future research to directly test the interactions between cannabis and aging.
Collapse
Affiliation(s)
- Hye Bin Yoo
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| | - Jennifer DiMuzio
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| | - Francesca M Filbey
- Center for BrainHealth, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
41
|
Neasta J, Darcq E, Jeanblanc J, Carnicella S, Ben Hamida S. GPCR and Alcohol-Related Behaviors in Genetically Modified Mice. Neurotherapeutics 2020; 17:17-42. [PMID: 31919661 PMCID: PMC7007453 DOI: 10.1007/s13311-019-00828-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest class of cell surface signaling receptors and regulate major neurobiological processes. Accordingly, GPCRs represent primary targets for the treatment of brain disorders. Several human genetic polymorphisms affecting GPCRs have been associated to different components of alcohol use disorder (AUD). Moreover, GPCRs have been reported to contribute to several features of alcohol-related behaviors in animal models. Besides traditional pharmacological tools, genetic-based approaches mostly aimed at deleting GPCR genes provided substantial information on how key GPCRs drive alcohol-related behaviors. In this review, we summarize the alcohol phenotypes that ensue from genetic manipulation, in particular gene deletion, of key GPCRs in rodents. We focused on GPCRs that belong to fundamental neuronal systems that have been shown as potential targets for the development of AUD treatment. Data are reviewed with particular emphasis on alcohol reward, seeking, and consumption which are behaviors that capture essential aspects of AUD. Literature survey indicates that in most cases, there is still a gap in defining the intracellular transducers and the functional crosstalk of GPCRs as well as the neuronal populations in which their signaling regulates alcohol actions. Further, the implication of only a few orphan GPCRs has been so far investigated in animal models. Combining advanced pharmacological technologies with more specific genetically modified animals and behavioral preclinical models is likely necessary to deepen our understanding in how GPCR signaling contributes to AUD and for drug discovery.
Collapse
Affiliation(s)
- Jérémie Neasta
- Laboratoire de Pharmacologie, Faculté de Pharmacie, University of Montpellier, 34093, Montpellier, France
| | - Emmanuel Darcq
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada
| | - Jérôme Jeanblanc
- Research Group on Alcohol and Pharmacodependences-INSERM U1247, University of Picardie Jules Verne, 80025, Amiens, France
| | - Sebastien Carnicella
- INSERM U1216, Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, 38000, Grenoble, France
| | - Sami Ben Hamida
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada.
| |
Collapse
|
42
|
Pavón FJ, Serrano A, Stouffer DG, Polis I, Roberto M, Cravatt BF, Martin-Fardon R, de Fonseca FR, Parsons LH. Ethanol-induced alterations in endocannabinoids and relevant neurotransmitters in the nucleus accumbens of fatty acid amide hydrolase knockout mice. Addict Biol 2019; 24:1204-1215. [PMID: 30421483 PMCID: PMC6551299 DOI: 10.1111/adb.12695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/24/2018] [Accepted: 10/12/2018] [Indexed: 01/05/2023]
Abstract
Deletion of fatty acid amide hydrolase (FAAH), enzyme responsible for degrading endocannabinoids, increases alcohol consumption and preference. However, there is a lack of data on neurochemical events in mice exposed to alcohol in the absence of FAAH. Extracellular levels of endocannabinoids and relevant neurotransmitters were measured by in vivo microdialysis in the nucleus accumbens (NAc) of FAAH knockout (KO) and wild-type (WT) mice during an ethanol (EtOH; 2 g/kg, ip) challenge in EtOH-naive and repeated (r) EtOH-treated mice. In both genotypes, EtOH treatment caused no changes in baseline endocannabinoid levels, although FAAH KO mice displayed higher baseline N-arachidonoylethanolamine levels than WT mice. EtOH challenge caused a sustained increase in 2-arachidonoylglycerol (2-AG) levels in EtOH-naive WT mice but not in FAAH KO mice. In contrast, 2-AG levels were decreased following EtOH challenge in (r)EtOH-treated mice in both genotypes. Whereas (r)EtOH-treated mice showed higher baseline dopamine and serotonin levels than EtOH-naive mice in WT mice, these differences were attenuated in FAAH KO mice. Significant differences in baseline γ-aminobutyric acid (GABA) and glutamate levels by EtOH history were observed in WT mice but not in FAAH KO mice. Moreover, opposed effects on glutamate response were observed after EtOH challenge in EtOH-naive and (r)EtOH-treated FAAH KO mice. Finally, FAAH deletion failed to show EtOH-induced locomotion sensitivity. These data provide evidence of a potential influence of 2-AG in the neurochemical response to EtOH exposure in the NAc.
Collapse
Affiliation(s)
- Francisco J. Pavón
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Antonia Serrano
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - David G. Stouffer
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Ilham Polis
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Rémi Martin-Fardon
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Loren H. Parsons
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
43
|
Giménez-Gómez P, Pérez-Hernández M, O'Shea E, Caso JR, Martín-Hernandez D, Cervera LA, Centelles MLGL, Gutiérrez-Lopez MD, Colado MI. Changes in brain kynurenine levels via gut microbiota and gut-barrier disruption induced by chronic ethanol exposure in mice. FASEB J 2019; 33:12900-12914. [PMID: 31509716 DOI: 10.1096/fj.201900491rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammatory processes have been shown to modify tryptophan (Trp) metabolism. Gut microbiota appears to play a significant role in the induction of peripheral and central inflammation. Ethanol (EtOH) exposure alters gut permeability, but its effects on Trp metabolism and the involvement of gut microbiota have not been studied. We analyzed several parameters of gut-barrier and of peripheral and central Trp metabolism following 2 different EtOH consumption patterns in mice, the binge model, drinking in the dark (DID), and the chronic intermittent (CI) consumption paradigm. Antibiotic treatment was used to evaluate gut microbiota involvement in the CI model. Mice exposed to CI EtOH intake, but not DID, show bacterial translocation and increased plasma LPS immediately after EtOH removal. Gut-barrier permeability to FITC-dextran is increased by CI, and, furthermore, intestinal epithelial tight-junction (TJ) disruption is observed (decreased expression of zonula occludens 1 and occludin) associated with increased matrix metalloproteinase (MMP)-9 activity and iNOS expression. CI EtOH, but not DID, increases kynurenine (Kyn) levels in plasma and limbic forebrain. Intestinal bacterial decontamination prevents the LPS increase but not the permeability to FITC-dextran, TJ disruption, or the increase in MMP-9 activity and iNOS expression. Although plasma Kyn levels are not affected by antibiotic treatment, the elevation of Kyn in brain is prevented, pointing to an involvement of microbiota in CI EtOH-induced changes in brain Trp metabolism. Additionally, CI EtOH produces depressive-like symptoms of anhedonia, which are prevented by the antibiotic treatment thus pointing to an association between anhedonia and the increase in brain Kyn and to the involvement of gut microbiota.-Giménez-Gómez, P., Pérez-Hernández, M., O'Shea, E., Caso, J. R., Martín-Hernández, D., Cervera, L. A., Centelles. M. L. G.-L., Gutiérrez-Lopez, M. D., Colado, M. I. Changes in brain kynurenine levels via gut microbiota and gut-barrier disruption induced by chronic ethanol exposure in mice.
Collapse
Affiliation(s)
- Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - David Martín-Hernandez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - Luis Alou Cervera
- Área de Microbiología, Departamento de Medicina, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | | | - María Dolores Gutiérrez-Lopez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Maria Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
44
|
Martin GG, Seeger DR, McIntosh AL, Milligan S, Chung S, Landrock D, Dangott LJ, Golovko MY, Murphy EJ, Kier AB, Schroeder F. Sterol Carrier Protein-2/Sterol Carrier Protein-x/Fatty Acid Binding Protein-1 Ablation Impacts Response of Brain Endocannabinoid to High-Fat Diet. Lipids 2019; 54:583-601. [PMID: 31487051 DOI: 10.1002/lipd.12192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
Brain endocannabinoids (EC) such as arachidonoylethanolamine (AEA) and 2-arachidonoylglycerol (2-AG) primarily originate from serum arachidonic acid (ARA), whose level is regulated in part by a cytosolic ARA-binding protein, that is, liver fatty acid binding protein-1 (FABP1), not expressed in the brain. Ablation of the Fabp1 gene (LKO) increases brain AEA and 2-AG by decreasing hepatic uptake of ARA to increase serum ARA, thereby increasing ARA availability for uptake by the brain. The brain also expresses sterol carrier protein-2 (SCP-2), which is also a cytosolic ARA-binding protein. To further resolve the role of SCP-2 independent of FABP1, mice ablated in the Scp-2/Scp-x gene (DKO) were crossed with mice ablated in the Fabp1 gene (LKO) mice to generate triple knock out (TKO) mice. TKO impaired the ability of LKO to increase brain AEA and 2-AG. While a high-fat diet (HFD) alone increased brain AEA, TKO impaired this effect. Overall, these TKO-induced blocks were not attributable to altered expression of brain proteins in ARA uptake, AEA/2-AG synthesis, or AEA/2-AG degrading enzymes. Instead, TKO reduced serum levels of free ARA and/or total ARA and thereby decreased ARA availability for uptake to the brain and downstream synthesis of AEA and 2-AG therein. In summary, Scp-2/Scp-x gene ablation in Fabp1 null (LKO) mice antagonized the impact of LKO and HFD on brain ARA and, subsequently, EC levels. Thus, both FABP1 and SCP-2 participate in regulating the EC system in the brain.
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Drew R Seeger
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Sherrelle Milligan
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Sarah Chung
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Lawrence J Dangott
- Protein Chemistry Laboratory, Texas A&M University, College Station, TX, 77843-2128, USA
| | - Mikhail Y Golovko
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Eric J Murphy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| |
Collapse
|
45
|
Ghosh S, O'Connell JF, Carlson OD, González‐Mariscal I, Kim Y, Moaddel R, Ghosh P, Egan JM. Linoleic acid in diets of mice increases total endocannabinoid levels in bowel and liver: modification by dietary glucose. Obes Sci Pract 2019; 5:383-394. [PMID: 31452923 PMCID: PMC6700518 DOI: 10.1002/osp4.344] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/17/2019] [Accepted: 04/28/2019] [Indexed: 12/15/2022] Open
Abstract
AIM Linoleic acid (LA) is an essential fatty acid involved in the biosynthesis of arachidonic acid and prostaglandins. LA is known to induce obesity and insulin resistance. In this study, two concentrations of LA with or without added glucose (G) were fed to mice to investigate their effects on endocannabinoid (EC) biology. MATERIALS AND METHODS Four groups of C57BL/6 mice were provided with diets containing 1% or 8% LA with or without added G (LAG) for 8 weeks. Body weights, food intake, circulating glucose and insulin levels were measured throughout the study. Following euthanasia, plasma, bowel and hepatic ECs, monoacylglycerol lipase and fatty acid amide hydroxylase protein levels (enzymes responsible for EC degradation) and transcriptional activity of PPARα in liver were quantified. Liver was probed for evidence of insulin receptor activity perturbation. RESULTS Increasing dietary LA from 1% to 8% significantly increased circulating, small bowel and hepatic ECs. 1%LAG fed mice had lowest feed efficiency, and only liver levels of both ECs were reduced by addition of G. Addition of G to 1% LA diets resulted in elevated monoacylglycerol lipase and fatty acid amide hydroxylase protein levels (p < 0.001 and p < 0.001, respectively) in liver due to increased transcriptional activity of PPARα (p < 0.05). The reduced EC levels with addition of G also correlated with a measure of enhanced insulin action. CONCLUSION In conclusion, body weight of mice is influenced by the source of calorie intake. Furthermore, tissue EC/g are dependent on tissue-specific synthesis and degradation that are modulated by dietary LA and G which also influence food efficiency, and down-stream insulin signalling pathways. The findings could potentially be useful information for weight management efforts in humans.
Collapse
Affiliation(s)
- S. Ghosh
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
- PharmacologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - J. F. O'Connell
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
| | - O. D. Carlson
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
| | - I. González‐Mariscal
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
| | - Y. Kim
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
| | - R. Moaddel
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
| | - P. Ghosh
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
| | - J. M. Egan
- National Institute on Aging, Laboratory of Clinical InvestigationNational Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
46
|
Abstract
Cannabinoid receptors, endocannabinoids and the enzymes responsible for their biosynthesis and degradation constitute the endocannabinoid system. In recent decades, the endocannabinoid system has attracted considerable interest as a potential therapeutic target in numerous pathological conditions. Its involvement in several physiological processes is well known, such as in energy balance, appetite stimulation, blood pressure, pain modulation, embryogenesis, nausea and vomiting control, memory, learning and immune response, among others, as well as in pathological conditions where it exerts a protective role in the development of certain disorders. As a result, it has been reported that changes in endocannabinoid levels may be related to neurological diseases such as Parkinson's disease, Huntington's disease, Alzheimer's disease and multiple sclerosis, as well as anorexia and irritable bowel syndrome. Alterations in the endocannabinoid system have also been associated with cancer, affecting the growth, migration and invasion of some tumours. Cannabinoids have been tested in several cancer types, including brain, breast and prostate cancers. Cannabinoids have shown promise as analgesics for the treatment of both inflammatory and neuropathic pain. There is also evidence for a role of the endocannabinoid system in the control of emotional states, and cannabinoids could prove useful in decreasing and palliating post-traumatic stress disorder symptoms and anxiolytic disorders. The role of the endocannabinoid system in addictions has also been examined, and cannabinoids have been postulated as alternative and co-adjuvant treatments in some abuse syndromes, mainly in ethanol and opioid abuses. The expression of the endocannabinoid system in the eye suggests that it could be a potential therapeutic target for eye diseases. Considering the importance of the endocannabinoid system and the therapeutic potential of cannabinoids in this vast number of medical conditions, several clinical studies with cannabinoid-based medications are ongoing. In addition, some cannabinoid-based medications have already been approved in various countries, including nabilone and dronabinol capsules for the treatment of nausea and vomiting associated with chemotherapy, dronabinol capsules for anorexia, an oral solution of dronabinol for both vomiting associated with chemotherapy and anorexia, a Δ9-tetrahydrocannabinol/cannabidiol oromucosal spray for pain related to cancer and for spasticity and pain associated with multiple sclerosis, and an oral solution of cannabidiol for Dravet and Lennox-Gastaut syndromes. Here, we review the available efficacy, safety and tolerability data for cannabinoids in a range of medical conditions.
Collapse
Affiliation(s)
- Ana Isabel Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramón y Cajal s/n, 28040 , Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramón y Cajal s/n, 28040 , Madrid, Spain. .,Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 , Madrid, Spain.
| |
Collapse
|
47
|
Basavarajappa BS, Joshi V, Shivakumar M, Subbanna S. Distinct functions of endogenous cannabinoid system in alcohol abuse disorders. Br J Pharmacol 2019; 176:3085-3109. [PMID: 31265740 DOI: 10.1111/bph.14780] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
Δ9 -tetrahydrocannabinol, the principal active component in Cannabis sativa extracts such as marijuana, participates in cell signalling by binding to cannabinoid CB1 and CB2 receptors on the cell surface. The CB1 receptors are present in both inhibitory and excitatory presynaptic terminals and the CB2 receptors are found in neuronal subpopulations in addition to microglial cells and astrocytes and are present in both presynaptic and postsynaptic terminals. Subsequent to the discovery of the endocannabinoid (eCB) system, studies have suggested that alcohol alters the eCB system and that this system plays a major role in the motivation to abuse alcohol. Preclinical studies have provided evidence that chronic alcohol consumption modulates eCBs and expression of CB1 receptors in brain addiction circuits. In addition, studies have further established the distinct function of the eCB system in the development of fetal alcohol spectrum disorders. This review provides a recent and comprehensive assessment of the literature related to the function of the eCB system in alcohol abuse disorders.
Collapse
Affiliation(s)
- Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,New York State Psychiatric Institute, New York, NY, USA.,Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Vikram Joshi
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Madhu Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| |
Collapse
|
48
|
Godlewski G, Cinar R, Coffey NJ, Liu J, Jourdan T, Mukhopadhyay B, Chedester L, Liu Z, Osei-Hyiaman D, Iyer MR, Park JK, Smith RG, Iwakura H, Kunos G. Targeting Peripheral CB 1 Receptors Reduces Ethanol Intake via a Gut-Brain Axis. Cell Metab 2019; 29:1320-1333.e8. [PMID: 31105045 PMCID: PMC6551287 DOI: 10.1016/j.cmet.2019.04.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/01/2019] [Accepted: 04/21/2019] [Indexed: 12/21/2022]
Abstract
Endocannabinoids acting on the cannabinoid-1 receptor (CB1R) or ghrelin acting on its receptor (GHS-R1A) both promote alcohol-seeking behavior, but an interaction between the two signaling systems has not been explored. Here, we report that the peripheral CB1R inverse agonist JD5037 reduces ethanol drinking in wild-type mice but not in mice lacking CB1R, ghrelin peptide or GHS-R1A. JD5037 treatment of alcohol-drinking mice inhibits the formation of biologically active octanoyl-ghrelin without affecting its inactive precursor desacyl-ghrelin. In ghrelin-producing stomach cells, JD5037 reduced the level of the substrate octanoyl-carnitine generated from palmitoyl-carnitine by increasing fatty acid β-oxidation. Blocking gastric vagal afferents abrogated the ability of either CB1R or GHS-R1A blockade to reduce ethanol drinking. We conclude that blocking CB1R in ghrelin-producing cells reduces alcohol drinking by inhibiting the formation of active ghrelin and its signaling via gastric vagal afferents. Thus, peripheral CB1R blockade may have therapeutic potential in the treatment of alcoholism.
Collapse
Affiliation(s)
- Grzegorz Godlewski
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nathan J Coffey
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jie Liu
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tony Jourdan
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bani Mukhopadhyay
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lee Chedester
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ziyi Liu
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Douglas Osei-Hyiaman
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Malliga R Iyer
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joshua K Park
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Roy G Smith
- Scripps Research Institute, Jupiter, FL 33458, USA
| | - Hiroshi Iwakura
- Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
49
|
Hodges EL, Ashpole NM. Aging circadian rhythms and cannabinoids. Neurobiol Aging 2019; 79:110-118. [PMID: 31035036 DOI: 10.1016/j.neurobiolaging.2019.03.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/11/2019] [Accepted: 03/17/2019] [Indexed: 01/04/2023]
Abstract
Numerous aspects of mammalian physiology exhibit cyclic daily patterns known as circadian rhythms. However, studies in aged humans and animals indicate that these physiological rhythms are not consistent throughout the life span. The simultaneous development of disrupted circadian rhythms and age-related impairments suggests a shared mechanism, which may be amenable to therapeutic intervention. Recently, the endocannabinoid system has emerged as a complex signaling network, which regulates numerous aspects of circadian physiology relevant to the neurobiology of aging. Agonists of cannabinoid receptor-1 (CB1) have consistently been shown to decrease neuronal activity, core body temperature, locomotion, and cognitive function. Paradoxically, several lines of evidence now suggest that very low doses of cannabinoids are beneficial in advanced age. One potential explanation for this phenomenon is that these drugs exhibit hormesis-a biphasic dose-response wherein low doses produce the opposite effects of higher doses. Therefore, it is important to determine the dose-, age-, and time-dependent effects of these substances on the regulation of circadian rhythms and other processes dysregulated in aging. This review highlights 3 fields-biological aging, circadian rhythms, and endocannabinoid signaling-to critically assess the therapeutic potential of endocannabinoid modulation in aged individuals. If the hormetic properties of exogenous cannabinoids are confirmed, we conclude that precise administration of these compounds may bidirectionally entrain central and peripheral circadian clocks and benefit multiple aspects of aging physiology.
Collapse
Affiliation(s)
- Erik L Hodges
- Pharmacology Division, Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, Oxford, MS, USA
| | - Nicole M Ashpole
- Pharmacology Division, Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, Oxford, MS, USA.
| |
Collapse
|
50
|
Turna J, Syan SK, Frey BN, Rush B, Costello MJ, Weiss M, MacKillop J. Cannabidiol as a Novel Candidate Alcohol Use Disorder Pharmacotherapy: A Systematic Review. Alcohol Clin Exp Res 2019; 43:550-563. [PMID: 30698831 DOI: 10.1111/acer.13964] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 01/20/2019] [Indexed: 01/23/2023]
Abstract
There is substantial interest in the therapeutic potential of cannabidiol (CBD), a nonpsychoactive cannabinoid found in plants of the genus Cannabis. The goal of the current systematic review was to characterize the existing literature on this topic and to evaluate the credibility of CBD as a candidate pharmacotherapy for alcohol use disorder (AUD). Using a comprehensive search strategy, 303 unique potential articles were identified and 12 ultimately met criteria for inclusion (8 using rodent models, 3 using healthy adult volunteers, and 1 using cell culture). In both rodent and cell culture models, CBD was found to exert a neuroprotective effect against adverse alcohol consequences on the hippocampus. In rodent models, CBD was found to attenuate alcohol-induced hepatotoxicity, specifically, alcohol-induced steatosis. Finally, findings from preclinical rodent models also indicate that CBD attenuates cue-elicited and stress-elicited alcohol seeking, alcohol self-administration, withdrawal-induced convulsions, and impulsive discounting of delayed rewards. In human studies, CBD was well tolerated and did not interact with the subjective effects of alcohol. Collectively, given its favorable effects on alcohol-related harms and addiction phenotypes in preclinical models, CBD appears to have promise as a candidate AUD pharmacotherapy. This is further bolstered by the absence of abuse liability and its general tolerability. A clear limitation to the literature is the paucity of human investigations. Human preclinical and clinical studies are needed to determine whether these positive effects in model systems substantively translate into clinically relevant outcomes.
Collapse
Affiliation(s)
- Jasmine Turna
- Michael G. DeGroote Centre for Medicinal Cannabis Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada.,Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Sabrina K Syan
- Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada.,Department of Psychology, Neuroscience, and Behavior, McMaster University, Hamilton, Ontario, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada.,Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Brian Rush
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Homewood Research Institute, Guelph, Ontario, Canada
| | | | - Mark Weiss
- Addiction Medicine Service, Homewood Health Centre, Guelph, Ontario, Canada
| | - James MacKillop
- Michael G. DeGroote Centre for Medicinal Cannabis Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada.,Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada.,Department of Psychology, Neuroscience, and Behavior, McMaster University, Hamilton, Ontario, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada.,Homewood Research Institute, Guelph, Ontario, Canada
| |
Collapse
|